1
|
Guo Y, Gujarati NA, Chow AK, Boysan BT, Bronstein R, He JC, Revelo MP, Pabla N, Rizzo RC, Das B, Mallipattu SK. A Small Molecule Agonist of Krüppel-Like Factor 15 in Proteinuric Kidney Disease. J Am Soc Nephrol 2024; 35:1671-1685. [PMID: 39133556 PMCID: PMC11617484 DOI: 10.1681/asn.0000000000000460] [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: 05/22/2024] [Accepted: 07/31/2024] [Indexed: 08/30/2024] Open
Abstract
Key Points A human podocyte-based high-throughput screen identified a novel agonist of Krüppel-like factor 15 (BT503), independent of glucocorticoid signaling. BT503 demonstrated renoprotective effects in three independent proteinuric kidney murine models. BT503 directly binds to inhibitor of nuclear factor kappa-B kinase subunit beta to inhibit NF-κB activation, which, subsequently restores Krüppel-like factor 15 under cell stress. Background Podocyte loss is the major driver of primary glomerular diseases such as FSGS. While systemic glucocorticoids remain the initial and primary therapy for these diseases, high-dose and chronic use of glucocorticoids is riddled with systemic toxicities. Krüppel-like factor 15 (KLF15) is a glucocorticoid-responsive gene, which is essential for the restoration of mature podocyte differentiation markers and stabilization of actin cytoskeleton in the setting of cell stress. Induction of KLF15 attenuates podocyte injury and glomerulosclerosis in the setting of cell stress. Methods A cell-based high-throughput screen with a subsequent structure–activity relationship study was conducted to identify novel agonists of KLF15 in human podocytes. Next, the agonist was tested in cultured human podocytes under cell stress and in three independent proteinuric models (LPS, nephrotoxic serum nephritis, and HIV-1 transgenic mice). A combination of RNA sequencing and molecular modeling with experimental validation was conducted to demonstrate the direct target of the agonist. Results The high-throughput screen with structure–activity relationship study identified BT503, a urea-based compound, as a novel agonist of KLF15, independent of glucocorticoid signaling. BT503 demonstrated protective effects in cultured human podocytes and in three independent proteinuric murine models. Subsequent molecular modeling with experimental validation shows that BT503 targets the inhibitor of nuclear factor kappa-B kinase complex by directly binding to inhibitor of nuclear factor kappa-B kinase subunit beta to inhibit canonical NF-κB signaling, which, in turn, restores KLF15 under cell stress, thereby rescuing podocyte loss and ameliorating kidney injury. Conclusions By developing and validating a cell-based high-throughput screen in human podocytes, we identified a novel agonist for KLF15 with salutary effects in proteinuric murine models through direct inhibition of inhibitor of nuclear factor kappa-B kinase subunit beta kinase activity.
Collapse
Affiliation(s)
- Yiqing Guo
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York
| | - Nehaben A. Gujarati
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York
| | - Andrew K. Chow
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York
| | - Brock T. Boysan
- Department of Chemistry, Stony Brook University, Stony Brook, New York
| | - Robert Bronstein
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York
| | - John C. He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Monica P. Revelo
- Department of Pathology, University of Utah, Salt Lake City, Utah
| | - Navjot Pabla
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, Ohio State University, Columbus, Ohio
| | - Robert C. Rizzo
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York
- Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, New York
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York
| | - Bhaskar Das
- Pharmaceutical Sciences, Long Island University, Brookville, New York
| | - Sandeep K. Mallipattu
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York
- Renal Section, Northport VA Medical Center, Northport, New York
| |
Collapse
|
2
|
Shih SY, Grant MP, Epure LM, Alad M, Lerouge S, Huk OL, Bergeron SG, Zukor DJ, Merle G, Im HJ, Antoniou J, Mwale F. Advances in the Regulation of Periostin for Osteoarthritic Cartilage Repair Applications. Biomolecules 2024; 14:1469. [PMID: 39595645 PMCID: PMC11592007 DOI: 10.3390/biom14111469] [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: 08/14/2024] [Revised: 09/17/2024] [Accepted: 09/17/2024] [Indexed: 11/28/2024] Open
Abstract
Emerging evidence indicates periostin (POSTN) is upregulated in patients with OA, and studies have shown that it can induce the activation of inflammatory cytokines and catabolic enzymes, making it a potential therapeutic target. Link N (LN) is a peptide fragment derived from the link protein and has been demonstrated as an anabolic-like factor and anti-catabolic and anti-inflammatory factors both in vitro and in vivo. This study aims to determine if LN can regulate POSTN expression and function in OA cartilage. Articular cartilage was recovered from donors undergoing total knee replacements to isolate chondrocytes and prepare osteochondral explants. Cells and explants were treated with POSTN and LN (1 and 100 μg) and measured for changes in POSTN expression and various matrix proteins, catabolic and proinflammatory factors, and signaling. To determine the effects of POSTN expression in vivo, a rabbit OA model was used. Immunoprecipitation and in silico modeling were used to determine peptide/POSTN interactions. Western blotting, PCR, and immunohistochemistry demonstrated that LN decreased POSTN expression both in vitro and in vivo. LN was also able to directly inhibit POSTN signaling in OA chondrocytes. In silico docking suggested the direct interaction of LN with POSTN at residues responsible for its oligomerization. Immunoprecipitation experiments confirmed the direct interaction of LN with POSTN and the destabilization of its oligomerization. This study demonstrates the ability of a peptide, LN, to suppress the overexpression and function of POSTN in OA cartilage.
Collapse
Affiliation(s)
- Sunny Y. Shih
- Department of Surgical and Interventional Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada (G.M.)
- Orthopaedic Research Laboratory, Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada
| | - Michael P. Grant
- Department of Surgical and Interventional Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada (G.M.)
- Orthopaedic Research Laboratory, Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada
| | - Laura M. Epure
- Department of Surgical and Interventional Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada (G.M.)
- Department of Orthopaedics, SMBD-Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
| | - Muskan Alad
- Department of Surgical and Interventional Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada (G.M.)
- Orthopaedic Research Laboratory, Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada
| | - Sophie Lerouge
- Department of Mechanical Engineering, École de Technologie Supérieure (ETS), Montreal, QC H3C 1K3, Canada
- Laboratory of Endovascular Biomaterials (LBeV), Centre de Recherche du CHUM (CRCHUM), Montreal, QC H2X 0A9, Canada
| | - Olga L. Huk
- Department of Surgical and Interventional Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada (G.M.)
- Department of Orthopaedics, SMBD-Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
| | - Stephane G. Bergeron
- Department of Surgical and Interventional Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada (G.M.)
- Department of Orthopaedics, SMBD-Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
| | - David J. Zukor
- Department of Surgical and Interventional Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada (G.M.)
- Department of Orthopaedics, SMBD-Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
| | - Géraldine Merle
- Department of Surgical and Interventional Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada (G.M.)
- Chemical Engineering Department, Polytechnique Montréal, Montreal, QC H3C 3A7, Canada
| | - Hee-Jeong Im
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
- Jesse Brown Veterans Affairs Medical Center (JBVAMC), Chicago, IL 60612, USA
| | - John Antoniou
- Department of Surgical and Interventional Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada (G.M.)
- Orthopaedic Research Laboratory, Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada
| | - Fackson Mwale
- Department of Surgical and Interventional Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada (G.M.)
- Orthopaedic Research Laboratory, Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada
| |
Collapse
|
3
|
Xiong X, Feng X, Ding Y. Periostin Induces Epithelial-Mesenchymal Transition via p38-MAPK Pathway in Human Renal Tubular Cells by High Glucose. Immun Inflamm Dis 2024; 12:e70077. [PMID: 39570100 PMCID: PMC11580286 DOI: 10.1002/iid3.70077] [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: 03/03/2024] [Revised: 10/22/2024] [Accepted: 11/06/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND Periostin mediates inflammation and fibrosis by regulating extracellular matrix adhesion, migration, and differentiation in multiple organ diseases. Studies have shown periostin mainly located in the dilated mesangium, tubulointerstitial and fibrotic regions of the diabetic kidney disease, which was negatively correlated with renal function. However, the underlying mechanism remains poorly explored. METHODS The expression of periostin in HK-2 cells was investigated under high glucose and high concentration of TGF-β1. The signaling pathway of periostin involved in epithelial-mesenchymal transdifferentiation of HK-2 cells was also validated. The expression of periostin were investigated by RT-PCR, western blot analysis and immunofluorescence assays with different concentrations of glucose and TGF-β1. The expression of E-Cad, α-SMA and p38 proteins were also detected. The effects of periostin, E-Cad, and α-SMA in high glucose were investigated by p38 inhibitors. To demonstrate the interaction among periostin, p38 and EMT markers, periostin under high glucose and high TGF-β1 was knocked down, resulting p38 and phosphorylated p38 was evaluated. RESULTS The combined of high glucose (HG, 22 mmol/L) and high TGF-β1 (10 ng/mL) upregulated the expression of periostin obviously, stimulating the expression of α-SMA and p38 while inhibiting the expression of E-Cad. p38 inhibitors reduced the expression of periostin and α-SMA while promoted E-Cad protein expression in HK-2 cells under HG conditions. Additionally, p38-MAPK signal pathway was involved in epithelial-mesenchymal transition of human renal tubules in high glucose environment. Significant, knockdown periostin expression effectively inhibited the expression of p38 and phosphorylated p38 under the combination of HG and high TGF-β1, verifying the interaction of periostin with the p38-MAPK signaling pathway. CONCLUSION Periostin, a downstream factor of TGF-β1, is positively regulated by TGF-β1 under HG condition, affecting the epithelial-interstitial differentiation of HK-2 cells via p38-MAPK signaling pathway. Therefore, periostin may serve as a biomarker of renal fibrosis in diabetic kidney disease.
Collapse
Affiliation(s)
- Xiaoling Xiong
- Department of Nephrology, Sir Run Run Shaw Hospital, College of MedicineZhejiang UniversityHangzhouChina
| | - Xing Feng
- Department of Thoracic Surgery, Affiliated Hangzhou First People's Hospital, School of MedicineWestlake UniversityHangzhouChina
| | - Yuqing Ding
- Department of Nephrology, Sir Run Run Shaw Hospital, College of MedicineZhejiang UniversityHangzhouChina
| |
Collapse
|
4
|
Sato T, Shizu R, Baba R, Ooka A, Hosaka T, Kanno Y, Yoshinari K. Pregnane X receptor inhibits the transdifferentiation of hepatic stellate cells by down-regulating periostin expression. Biochem J 2024; 481:1173-1186. [PMID: 39171361 DOI: 10.1042/bcj20240172] [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: 04/14/2024] [Revised: 08/07/2024] [Accepted: 08/21/2024] [Indexed: 08/23/2024]
Abstract
Pregnane X receptor (PXR) is a xenobiotic-sensing nuclear receptor that plays a key role in drug metabolism. Recently, PXR was found to attenuate the development of liver cancer by suppressing epithelial-mesenchymal transition (EMT) in liver cancer cells in a mouse model of two-stage chemical carcinogenesis. To elucidate the role of PXR in the EMT of liver cancer cells, we focused on its role in hepatic stellate cells (HSCs), which are components of the tumor microenvironment in hepatocellular carcinoma (HCC). Human HSC-derived LX-2 cells stably expressed destabilization domain (DD)-fused human PXR (hPXR-LX2 cells). Human HCC-derived HepG2 cells were transfected with the EMT marker VIM promoter-regulated reporter plasmid and co-cultured with hPXR-LX2 cells or treated with hPXR-LX2-derived conditioned medium (CM). Co-culture or CM treatment increased reporter activity in HepG2 cells. This induction was attenuated upon PXR activation in hPXR-LX2 cells by treatment with the DD-stabilizing chemical Shield-1 and the human PXR ligand rifampicin. PXR activation in hPXR-LX2 cells exhibited inhibition of TGF-β1-induced transdifferentiation, supported by observations of morphological changes and protein or mRNA levels of the transdifferentiation markers COL1A1 and FN1. PXR activation in hPXR-LX2 cells also attenuated the mRNA levels of the key transdifferentiation factor, POSTN. Treatment of hPXR-LX2 cells with recombinant POSTN restored the PXR-mediated suppression of transdifferentiation. Reporter assays with the POSTN promoter showed that PXR inhibited the NF-κB-mediated transcription of POSTN. Consequently, PXR activation in HSCs is expected to inhibit transdifferentiation by down-regulating POSTN expression, thereby suppressing EMT of liver cancer cells.
Collapse
Affiliation(s)
- Takumi Sato
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526 Japan
| | - Ryota Shizu
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526 Japan
| | - Ryonosuke Baba
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526 Japan
| | - Akira Ooka
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526 Japan
| | - Takuomi Hosaka
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526 Japan
| | - Yuichiro Kanno
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526 Japan
| | - Kouichi Yoshinari
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526 Japan
| |
Collapse
|
5
|
Hong YK, Hwang DY, Yang CC, Cheng SM, Chen PC, Aala WJ, I-Chen Harn H, Evans ST, Onoufriadis A, Liu SL, Lin YC, Chang YH, Lo TK, Hung KS, Lee YC, Tang MJ, Lu KQ, McGrath JA, Hsu CK. Profibrotic Subsets of SPP1 + Macrophages and POSTN + Fibroblasts Contribute to Fibrotic Scarring in Acne Keloidalis. J Invest Dermatol 2024; 144:1491-1504.e10. [PMID: 38218364 DOI: 10.1016/j.jid.2023.12.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/05/2023] [Accepted: 12/19/2023] [Indexed: 01/15/2024]
Abstract
Acne keloidalis is a primary scarring alopecia characterized by longstanding inflammation in the scalp causing keloid-like scar formation and hair loss. Histologically, acne keloidalis is characterized by mixed leukocytic infiltrates in the acute stage followed by a granulomatous reaction and extensive fibrosis in the later stages. To further explore its pathogenesis, bulk RNA sequencing, single-cell RNA sequencing, and spatial transcriptomics were applied to occipital scalp biopsy specimens of lesional and adjacent no-lesional skin in patients with clinically active disease. Unbiased clustering revealed 19 distinct cell populations, including 2 notable populations: POSTN+ fibroblasts with enriched extracellular matrix signatures and SPP1+ myeloid cells with an M2 macrophage phenotype. Cell communication analyses indicated that fibroblasts and myeloid cells communicated by SPP1 signaling networks in lesional skin. A reverse transcriptomics in silico approach identified corticosteroids as possessing the capability to reverse the gene expression signatures of SPP1+ myeloid cells and POSTN+ fibroblasts. Intralesional corticosteroid injection greatly reduced SPP1 and POSTN gene expression as well as acne keloidalis disease activity. Spatial transcriptomics and immunofluorescence staining verified microanatomic specificity of SPP1+ myeloid cells and POSTN+ fibroblasts with disease activity. In summary, the communication between POSTN+ fibroblasts and SPP1+ myeloid cells by SPP1 axis may contribute to the pathogenesis of acne keloidalis.
Collapse
Affiliation(s)
- Yi-Kai Hong
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan
| | - Daw-Yang Hwang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Chao-Chun Yang
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan
| | - Siao Muk Cheng
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Peng-Chieh Chen
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wilson Jr Aala
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hans I-Chen Harn
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Spencer T Evans
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Alexandros Onoufriadis
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, London, United Kingdom
| | - Si-Lin Liu
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Chen Lin
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Han Chang
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tzu-Kun Lo
- Department of Dermatology, Tainan Municipal An-Nan Hospital, Tainan, Taiwan
| | - Kuo-Shu Hung
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Chao Lee
- PhD Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Ming-Jer Tang
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan; Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kurt Q Lu
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - John A McGrath
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; St John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London, London, United Kingdom
| | - Chao-Kai Hsu
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan; Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
6
|
Li X, Shan J, Chen X, Cui H, Wen G, Yu Y. Decellularized diseased tissues: current state-of-the-art and future directions. MedComm (Beijing) 2023; 4:e399. [PMID: 38020712 PMCID: PMC10661834 DOI: 10.1002/mco2.399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/04/2023] [Accepted: 09/12/2023] [Indexed: 12/01/2023] Open
Abstract
Decellularized matrices derived from diseased tissues/organs have evolved in the most recent years, providing novel research perspectives for understanding disease occurrence and progression and providing accurate pseudo models for developing new disease treatments. Although decellularized matrix maintaining the native composition, ultrastructure, and biomechanical characteristics of extracellular matrix (ECM), alongside intact and perfusable vascular compartments, facilitates the construction of bioengineered organ explants in vitro and promotes angiogenesis and tissue/organ regeneration in vivo, the availability of healthy tissues and organs for the preparation of decellularized ECM materials is limited. In this paper, we review the research advancements in decellularized diseased matrices. Considering that current research focuses on the matrices derived from cancers and fibrotic organs (mainly fibrotic kidney, lungs, and liver), the pathological characterizations and the applications of these diseased matrices are mainly discussed. Additionally, a contrastive analysis between the decellularized diseased matrices and decellularized healthy matrices, along with the development in vitro 3D models, is discussed in this paper. And last, we have provided the challenges and future directions in this review. Deep and comprehensive research on decellularized diseased tissues and organs will promote in-depth exploration of source materials in tissue engineering field, thus providing new ideas for clinical transformation.
Collapse
Affiliation(s)
- Xiang Li
- Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jianyang Shan
- Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xin Chen
- Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
- College of Fisheries and Life ScienceShanghai Ocean UniversityShanghaiChina
| | - Haomin Cui
- Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Gen Wen
- Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yaling Yu
- Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
- Institute of Microsurgery on ExtremitiesShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
7
|
Duan X, Chen C, Liu X, Wang T, Feng S, Li J, Li G. Interference of periostin attenuates pathological changes, proinflammatory markers and renal fibrosis in diabetic kidney injury. Genes Genomics 2023; 45:1389-1397. [PMID: 37248423 DOI: 10.1007/s13258-023-01400-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 05/11/2023] [Indexed: 05/31/2023]
Abstract
BACKGROUND Diabetic nephropathy (DN) is a prevalent complication of diabetes, in which inflammation and fibrosis are the significant pathogenesis. Periostin is a matricellular protein that functions on stabilizing the extracellular matrix by binding to integrins during development. This study aimed to explored the role of periostin in DN. METHODS The animal and cell models of DN were constructed in streptozocin (STZ)-induced mice and high glucose-challenged human mesangial cells (HMCs). The role of periostin in pathological changes, inflammation and fibrosis in DN was investigated through biochemical detection, HE and Masson staining and scores, western blot, enzyme‑linked immunosorbent assay (ELISA) and real-time quantitative PCR (RT-qPCR) assays. RESULTS Knockdown of periostin counteracted the STZ-induced the ratio of kidney weight and body weight, and the concentrations of urine albumin excretion (UAE), serum creatinine (Scr), urine albumin/creatinine ratio (UACR) and blood urea nitrogen (BUN) in mice. Moreover, silencing of periostin alleviated the pathological manifestations and reduced the concentrations of IL-6, TNF-α and IL-1β in mice kidney tissues and sera. Also, downregulation of periostin decreased the relative protein expression of fibronectin, collagen IV and α-SMA in kidney tissues. Meanwhile, interference of periostin attenuated the levels of pro-inflammation factors and the expressions of fibrosis markers in HG-induced HMCs. CONCLUSION Interference of periostin resisted DN via attenuating the pro-inflammatory cytokines release and renal fibrosis in diabetic kidney injury. Our study establishes a basis for its further study and underlying application in clinical practice in diagnosing and treating diabetic kidney injury or other relevant diseases.
Collapse
Affiliation(s)
- Xiaoting Duan
- Department of Nephrology, Affiliated Hospital of Hebei Engineering University, No. 81 Congtai Road, Handan City, Hebei Province056000, China
| | - Cheng Chen
- The Second Department of Oncology, Affiliated Hospital of Hebei Engineering University, Hebei, 056000, China
| | - Xiaoli Liu
- Department of Nephrology, Affiliated Hospital of Hebei Engineering University, No. 81 Congtai Road, Handan City, Hebei Province056000, China
| | - Taoxia Wang
- Department of Nephrology, Affiliated Hospital of Hebei Engineering University, No. 81 Congtai Road, Handan City, Hebei Province056000, China
| | - Shuning Feng
- Department of Nephrology, Affiliated Hospital of Hebei Engineering University, No. 81 Congtai Road, Handan City, Hebei Province056000, China
| | - Jianwei Li
- Department of Nephrology, Affiliated Hospital of Hebei Engineering University, No. 81 Congtai Road, Handan City, Hebei Province056000, China
| | - Guiying Li
- Department of Nephrology, Affiliated Hospital of Hebei Engineering University, No. 81 Congtai Road, Handan City, Hebei Province056000, China.
| |
Collapse
|
8
|
The Multiple Roles of Periostin in Non-Neoplastic Disease. Cells 2022; 12:cells12010050. [PMID: 36611844 PMCID: PMC9818388 DOI: 10.3390/cells12010050] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/05/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Periostin, identified as a matricellular protein and an ECM protein, plays a central role in non-neoplastic diseases. Periostin and its variants have been considered to be normally involved in the progression of most non-neoplastic diseases, including brain injury, ocular diseases, chronic rhinosinusitis, allergic rhinitis, dental diseases, atopic dermatitis, scleroderma, eosinophilic esophagitis, asthma, cardiovascular diseases, lung diseases, liver diseases, chronic kidney diseases, inflammatory bowel disease, and osteoarthrosis. Periostin interacts with protein receptors and transduces signals primarily through the PI3K/Akt and FAK two channels as well as other pathways to elicit tissue remodeling, fibrosis, inflammation, wound healing, repair, angiogenesis, tissue regeneration, bone formation, barrier, and vascular calcification. This review comprehensively integrates the multiple roles of periostin and its variants in non-neoplastic diseases, proposes the utility of periostin as a biological biomarker, and provides potential drug-developing strategies for targeting periostin.
Collapse
|
9
|
Pană N, Căpușă C. Periostin as a Biomarker in the Setting of Glomerular Diseases-A Review of the Current Literature. Biomedicines 2022; 10:biomedicines10123211. [PMID: 36551967 PMCID: PMC9775428 DOI: 10.3390/biomedicines10123211] [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: 10/24/2022] [Revised: 11/20/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Chronic kidney disease (CKD) is a highly prevalent and potential progressive condition with life-threatening consequences. Glomerular diseases (glomerulopathies) are causes of CKD that are potentially amenable by specific therapies. Significant resources have been invested in the identification of novel biomarkers of CKD progression and new targets for treatment. By using experimental models of kidney diseases, periostin has been identified amongst the most represented matricellular proteins that are commonly involved in the inflammation and fibrosis that characterize progressive kidney diseases. Periostin is highly expressed during organogenesis, with scarce expression in mature healthy tissues, but it is upregulated in multiple disease settings characterized by tissue injury and remodeling. Periostin was the most highly expressed matriceal protein in both animal models and in patients with glomerulopathies. Given that periostin is readily secreted from injury sites, and the variations in its humoral levels compared to the normal state were easily detectable, its potential role as a biomarker is suggested. Moreover, periostin expression was correlated with the degree of histological damage and with kidney function decline in patients with CKD secondary to both inflammatory (IgA nephropathy) and non-inflammatory (membranous nephropathy) glomerulopathies, while also displaying variability secondary to treatment response. The scope of this review is to summarize the existing evidence that supports the role of periostin as a novel biomarker in glomerulopathies.
Collapse
Affiliation(s)
- Nicolae Pană
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Diaverum Morarilor Clinic of Nephrology and Dialysis, 022452 Bucharest, Romania
| | - Cristina Căpușă
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- "Dr Carol Davila" Teaching Hospital of Nephrology, 010731 Bucharest, Romania
| |
Collapse
|
10
|
Regulatory Networks, Management Approaches, and Emerging Treatments of Nonalcoholic Fatty Liver Disease. Can J Gastroenterol Hepatol 2022; 2022:6799414. [PMID: 36397950 PMCID: PMC9666027 DOI: 10.1155/2022/6799414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/05/2022] [Indexed: 11/09/2022] Open
Abstract
The pathogenesis of NAFLD is complex and diverse, involving multiple signaling pathways and cytokines from various organs. Hepatokines, stellakines, adipokines, and myokines secreted by hepatocytes, hepatic stellate cells, adipose tissue, and myocytes play an important role in the occurrence and development of nonalcoholic fatty liver disease (NAFLD). The nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) contributes to the progression of NAFLD by mediating liver inflammation, immune response, hepatocyte death, and later compensatory proliferation. In this review, we first discuss the crosstalk and interaction between hepatokines, stellakines, adipokines, and myokines and NF-κB in NAFLD. The characterization of the crosstalk of NF-κB with these factors will provide a better understanding of the molecular mechanisms involved in the progression of NAFLD. In addition, we examine new expert management opinions for NAFLD and explore the therapeutic potential of silymarin in NAFLD/NASH.
Collapse
|
11
|
Blocking Periostin Prevented Development of Inflammation in Rhabdomyolysis-Induced Acute Kidney Injury Mice Model. Cells 2022; 11:cells11213388. [DOI: 10.3390/cells11213388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/13/2022] [Accepted: 10/20/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Rhabdomyolysis is the collapse of damaged skeletal muscle and the leakage of muscle-cell contents, such as electrolytes, myoglobin, and other sarcoplasmic proteins, into the circulation. The glomeruli filtered these products, leading to acute kidney injury (AKI) through several mechanisms, such as intratubular obstruction secondary to protein precipitation. The prognosis is highly mutable and depends on the underlying complications and etiologies. New therapeutic plans to reduce AKI are now needed. Up to now, several cellular pathways, with the nuclear factor kappa beta (NF-kB), as well as the proinflammatory effects on epithelial and tubular epithelial cells, have been recognized as the major pathway for the initiation of the matrix-producing cells in AKI. Recently, it has been mentioned that periostin (POSTN), an extracellular matrix protein, is involved in the development of inflammation through the modulation of the NF-kB pathway. However, how POSTN develops the inflammation protection in AKI by rhabdomyolysis is uncertain. This study aimed to investigate the role of POSTN in a rhabdomyolysis mice model of AKI induced by an intramuscular injection of 50% glycerol. Methods: In vivo, we performed an intramuscular injection of 50% glycerol (5 mg/kg body weight) to make rhabdomyolysis-induced AKI. We examined the expression level of POSTN through the progression of AKI after glycerol intramuscular injection for C57BL/6J wildtype (WT) mice. We sacrificed mice at 72 h after glycerol injection. We made periostin-null mice to examine the role of POSTN in acute renal failure. The role of periostin was further examined through in vitro methods. The development of renal inflammation is linked with the NF-kB pathway. To examine the POSTN function, we administrated hemin (100 μM) on NIH-3T3 fibroblast cells, and the following signaling pathways were examined. Results: The expression of periostin was highly increased, peaking at about 72 h after glycerol injection. The expression of inflammation-associated mRNAs such as monocyte chemotactic protein-1 (MCP-1), tumor necrosis factor-alpha (TNF-a) and IL-6, and tubular injury score in H-E staining were more reduced in POSTN-null mice than WT mice at 72 h after glycerol injection. Conclusion: POSTN was highly expressed in the kidney through rhabdomyolysis and was a positive regulator of AKI. Targeting POSTN might propose a new therapeutic strategy against the development of acute renal failure.
Collapse
|
12
|
Sari A, Dogan S, Nibali L, Koseoglu S. Evaluation of IL-23p19/Ebi3 (IL-39) gingival crevicular fluid levels in periodontal health, gingivitis, and periodontitis. Clin Oral Investig 2022; 26:7209-7218. [PMID: 35986791 DOI: 10.1007/s00784-022-04681-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 08/10/2022] [Indexed: 11/28/2022]
Abstract
Abstract
Objectives
IL-23p19/Ebi3 (IL-39) was described as a new IL-12 family member. The aim of this study is to evaluate the gingival crevicular fluid (GCF) IL-39 levels in periodontal diseases and health and to correlate them to GCF levels of IL-1β and periostin.
Materials and methods
Sixty-six adult patients were included in the study. The study design was comprised of three groups, each containing 22 individuals: the periodontally healthy (PH), gingivitis (G), and periodontitis (P) groups. The clinical periodontal parameters were recorded and GCF samples were collected from the participants. GCF interleukin (IL)-39, IL-1β, and periostin levels were examined using the enzyme-linked immunosorbent assay.
Results
GCF IL‑1β, periostin, and IL-39 levels were higher in the P and G groups than in the PH group (p < 0.001). Positive correlations were detected between all GCF biochemical parameters and clinical periodontal parameters (p < 0.05). In the multivariate generalized linear regression analysis, the P (β = 37.6, 95% CI = 22.9–52.4) and G (β = 28.4, 95% CI = 15.8–41) groups were associated with GCF IL-39 levels (p < 0.001).
Conclusion
IL-39 levels were elevated in the presence of periodontal disease paralleling the increase in IL‑1β and periostin levels. IL-39 may have a role in the periodontal inflammation process.
Statement of clinical relevance
IL-39, a new cytokine from the IL-12 family, can be a possible predictor marker of periodontal diseases.
Collapse
|
13
|
Abbad L, Prakoura N, Michon A, Chalghoumi R, Reichelt-Wurm S, Banas MC, Chatziantoniou C. Role of Periostin and Nuclear Factor-κB Interplay in the Development of Diabetic Nephropathy. Cells 2022; 11:cells11142212. [PMID: 35883655 PMCID: PMC9320904 DOI: 10.3390/cells11142212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 02/06/2023] Open
Abstract
Diabetic nephropathy (DN) remains the most common reason for end-stage renal disease and a leading cause of kidney replacement therapy. Multifactorial pathophysiological mechanisms underlie the development of DN. Among the signalling pathways involved, nuclear factor-κB (NF-κB) plays a key role in pathogenesis triggering inflammation, oxidative stress and fibrosis. Recent evidence shows that periostin, a matricellular protein, is involved in the development of renal glomerular diseases through interaction with NF-κB signalling. The aim of the present study is to investigate the contribution of periostin and its interaction with NF-κB in DN development. To this end, we used the BTBR ob/ob mice model of diabetes type 2, and we applied transcriptomic analysis, immunostaining and methods quantifying protein and mRNA expressions. We found that increased periostin expression was correlated with decreased renal function, advanced stage renal damage and fibrosis, and NF-κB activation. Subsequently, we identified novel pathways and genes regulated by the NF-κB-periostin interaction which are involved in the mechanisms of progression of DN. Some of these genes, such as FGF1 and GDF15, have the potential to be new biomarkers and/or targets for the therapy of DN.
Collapse
Affiliation(s)
- Lilia Abbad
- Unite Mixte de Recherche Scientific 1155, Institut National de la Sante et de la Recherche Medicale, Tenon Hospital, 75020 Paris, France; (L.A.); (N.P.); (A.M.); (R.C.)
- Faculty of Medicine, Sorbonne University, 75020 Paris, France
| | - Niki Prakoura
- Unite Mixte de Recherche Scientific 1155, Institut National de la Sante et de la Recherche Medicale, Tenon Hospital, 75020 Paris, France; (L.A.); (N.P.); (A.M.); (R.C.)
- Faculty of Medicine, Sorbonne University, 75020 Paris, France
| | - Arthur Michon
- Unite Mixte de Recherche Scientific 1155, Institut National de la Sante et de la Recherche Medicale, Tenon Hospital, 75020 Paris, France; (L.A.); (N.P.); (A.M.); (R.C.)
- Faculty of Medicine, Sorbonne University, 75020 Paris, France
| | - Rym Chalghoumi
- Unite Mixte de Recherche Scientific 1155, Institut National de la Sante et de la Recherche Medicale, Tenon Hospital, 75020 Paris, France; (L.A.); (N.P.); (A.M.); (R.C.)
- Faculty of Medicine, Sorbonne University, 75020 Paris, France
| | - Simone Reichelt-Wurm
- Department of Nephrology, University Hospital Regensburg, D-93053 Regensburg, Germany; (S.R.-W.); (M.C.B.)
| | - Miriam C. Banas
- Department of Nephrology, University Hospital Regensburg, D-93053 Regensburg, Germany; (S.R.-W.); (M.C.B.)
| | - Christos Chatziantoniou
- Unite Mixte de Recherche Scientific 1155, Institut National de la Sante et de la Recherche Medicale, Tenon Hospital, 75020 Paris, France; (L.A.); (N.P.); (A.M.); (R.C.)
- Faculty of Medicine, Sorbonne University, 75020 Paris, France
- Correspondence:
| |
Collapse
|
14
|
The fibrogenic niche in kidney fibrosis: components and mechanisms. Nat Rev Nephrol 2022; 18:545-557. [PMID: 35788561 DOI: 10.1038/s41581-022-00590-z] [Citation(s) in RCA: 168] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2022] [Indexed: 02/08/2023]
Abstract
Kidney fibrosis, characterized by excessive deposition of extracellular matrix (ECM) that leads to tissue scarring, is the final common outcome of a wide variety of chronic kidney diseases. Rather than being distributed uniformly across the kidney parenchyma, renal fibrotic lesions initiate at certain focal sites in which the fibrogenic niche is formed in a spatially confined fashion. This niche provides a unique tissue microenvironment that is orchestrated by a specialized ECM network consisting of de novo-induced matricellular proteins. Other structural elements of the fibrogenic niche include kidney resident and infiltrated inflammatory cells, extracellular vesicles, soluble factors and metabolites. ECM proteins in the fibrogenic niche recruit soluble factors including WNTs and transforming growth factor-β from the extracellular milieu, creating a distinctive profibrotic microenvironment. Studies using decellularized ECM scaffolds from fibrotic kidneys show that the fibrogenic niche autonomously promotes fibroblast proliferation, tubular injury, macrophage activation and endothelial cell depletion, pathological features that recapitulate key events in the pathogenesis of chronic kidney disease. The concept of the fibrogenic niche represents a paradigm shift in understanding of the mechanism of kidney fibrosis that could lead to the development of non-invasive biomarkers and novel therapies not only for chronic kidney disease, but also for fibrotic diseases of other organs.
Collapse
|
15
|
Gao F, Bai R, Qin W, Liang B, Yang Z, Yang H. Angiotensin II induces the expression of periostin to promote foam cell formation in oxLDL-treated macrophages. Int J Cardiol 2022; 347:46-53. [PMID: 34793856 DOI: 10.1016/j.ijcard.2021.11.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 09/21/2021] [Accepted: 11/07/2021] [Indexed: 11/05/2022]
Abstract
A matricellular protein periostin has been documented to promote macrophage recruitment in atherosclerotic lesions. However, the role of periostin in macrophage foam cell formation is still unknown. In this study, we examined the expression and function of periostin in cholesterol homeostasis in macrophages. The role of periostin in mediating Ang II-induced foam cell formation was also investigated. The mechanism by which Ang II induced the expression of periostin was explored. It was found that oxLDL treatment significantly increased the expression and secretion of periostin in THP-1 macrophages. Knockdown of periostin blocked oxLDL-induced lipid accumulation and enhanced cholesterol efflux. In contrast, treatment with recombinant periostin protein enhanced oxLDL-induced macrophage foam cell formation. Ang II caused a time-dependent induction of periostin in THP-1 macrophages, which was ascribed to Twist2-mediated transactivation of periostin. Ang II treatment significantly augmented lipid accumulation in THP-1 macrophages, and knockdown of periostin blocked the effect of Ang II on foam cell formation. Moreover, periostin depletion restored cholesterol efflux in Ang II-treated THP-1 macrophages. Clinically, there was a significant positive correlation between serum periostin and Ang II levels in patients with atherosclerosis. Collectively, we show that periostin is essential for Ang II-induced enhancement of macrophage foam cell formation via suppression of cholesterol efflux.
Collapse
Affiliation(s)
- Fen Gao
- Departments of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Rui Bai
- Departments of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Weiwei Qin
- Departments of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Bin Liang
- Departments of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Zhiming Yang
- Departments of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Huiyu Yang
- Departments of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
16
|
Jang SY, Kim J, Park JT, Liu CY, Korn BS, Kikkawa DO, Lee EJ, Yoon JS. Therapeutic Potential of Targeting Periostin in the Treatment of Graves' Orbitopathy. Front Endocrinol (Lausanne) 2022; 13:900791. [PMID: 35707463 PMCID: PMC9189304 DOI: 10.3389/fendo.2022.900791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Periostin is a matricellular protein that is ubiquitously expressed in normal human tissues and is involved in pathologic mechanism of chronic inflammatory and fibrotic disease. In this study we investigate periostin in the pathogenesis of Graves' orbitopathy (GO) using human orbital adipose tissue obtained from surgery and primary cultured orbital fibroblasts in vitro. POSTN (gene encoding periostin) expression in Graves' orbital tissues and healthy control tissues was studied, and the role of periostin in GO pathologic mechanism was examined through small-interfering RNA (siRNA)-mediated silencing. POSTN gene expression was significantly higher in Graves' orbital tissues than healthy control tissues in real-time PCR results, and immunohistochemical staining revealed higher expression of periostin in Graves' orbital tissues than normal tissues. Silencing periostin using siRNA transfection significantly attenuated TGF-β-induced profibrotic protein production and phosphorylated p38 and SMAD protein production. Knockdown of periostin inhibited interleukin-1 β -induced proinflammatory cytokines production as well as phosphorylation of NF-κB and Ak signaling protein. Adipocyte differentiation was also suppressed in periostin-targeting siRNA transfected GO cells. We hypothesize that periostin contributes to the pathogenic process of inflammation, fibrosis and adipogenesis of GO. Our study provides in vitro evidence that periostin may be a novel potential therapeutic target for the treatment of GO.
Collapse
Affiliation(s)
- Sun Young Jang
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, South Korea
| | - Jinjoo Kim
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Jung Tak Park
- Department of Internal Medicine, College of Medicine, Institute of Kidney Disease Research, Yonsei University, Seoul, South Korea
| | - Catherine Y. Liu
- Division of Oculofacial Plastic and Reconstructive Surgery, University of California, San Diego, La Jolla, CA, United States
| | - Bobby S. Korn
- Division of Oculofacial Plastic and Reconstructive Surgery, University of California, San Diego, La Jolla, CA, United States
| | - Don O. Kikkawa
- Division of Oculofacial Plastic and Reconstructive Surgery, University of California, San Diego, La Jolla, CA, United States
| | - Eun Jig Lee
- Division of Endocrinology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Jin Sook Yoon
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, South Korea
- *Correspondence: Jin Sook Yoon,
| |
Collapse
|
17
|
The Usefulness of Urinary Periostin, Cytokeratin-18, and Endoglin for Diagnosing Renal Fibrosis in Children with Congenital Obstructive Nephropathy. J Clin Med 2021; 10:jcm10214899. [PMID: 34768419 PMCID: PMC8585114 DOI: 10.3390/jcm10214899] [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: 09/19/2021] [Revised: 10/11/2021] [Accepted: 10/20/2021] [Indexed: 12/28/2022] Open
Abstract
Congenital obstructive nephropathy (CON) leads to renal fibrosis and chronic kidney disease. The aim of the study was to investigate the predictive value of urinary endoglin, periostin, cytokeratin-18, and transforming growth factor-β1 (TGF-β1) for assessing the severity of renal fibrosis in 81 children with CON and 60 controls. Children were divided into three subgroups: severe, moderate scars, and borderline lesions based on 99mTc-ethylenedicysteine scintigraphy results. Periostin, periostin/Cr, and cytokeratin-18 levels were significantly higher in the study group compared to the controls. Children with severe scars had significantly higher urinary periostin/Cr levels than those with borderline lesions. In multivariate analysis, only periostin and cytokeratin-18 were independently related to the presence of severe and moderate scars, and periostin was independently related to borderline lesions. However, periostin did not differentiate advanced scars from borderline lesions. In ROC analysis, periostin and periostin/Cr demonstrated better diagnostic profiles for detection of advanced scars than TGF-β1 and cytokeratin-18 (AUC 0.849; 0.810 vs. 0.630; 0.611, respectively) and periostin for detecting borderline lesions than endoglin and periostin/Cr (AUC 0.777 vs. 0.661; 0.658, respectively). In conclusion, periostin seems to be a promising, non-invasive marker for assessing renal fibrosis in children with CON. CK-18 and TGF-β1 demonstrated low utility, and endoglin was not useful for diagnosing advanced scars.
Collapse
|
18
|
Möller-Hackbarth K, Dabaghie D, Charrin E, Zambrano S, Genové G, Li X, Wernerson A, Lal M, Patrakka J. Retinoic acid receptor responder1 promotes development of glomerular diseases via the Nuclear Factor-κB signaling pathway. Kidney Int 2021; 100:809-823. [PMID: 34147551 DOI: 10.1016/j.kint.2021.05.036] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 05/11/2021] [Accepted: 05/20/2021] [Indexed: 12/20/2022]
Abstract
Inflammatory pathways are activated in most glomerular diseases but molecular mechanisms driving them in kidney tissue are poorly known. We identified retinoic acid receptor responder 1 (Rarres1) as a highly podocyte-enriched protein in healthy kidneys. Studies in podocyte-specific knockout animals indicated that Rarres1 was not needed for the normal development or maintenance of the glomerulus filtration barrier and did not modulate the outcome of kidney disease in a model of glomerulonephritis. Interestingly, we detected an induction of Rarres1 expression in glomerular and peritubular capillary endothelial cells in IgA and diabetic kidney disease, as well as in ANCA-associated vasculitis. Analysis of publicly available RNA data sets showed that the induction of Rarres1 expression was a common molecular mechanism in chronic kidney diseases. A conditional knock-in mouse line, overexpressing Rarres1 specifically in endothelial cells, did not show any obvious kidney phenotype. However, the overexpression promoted the progression of kidney damage in a model of glomerulonephritis. In line with this, conditional knock-out mice, lacking Rarres1 in endothelial cells, were partially protected in the disease model. Mechanistically, Rarres1 promoted inflammation and fibrosis via transcription factor Nuclear Factor-κB signaling pathway by activating receptor tyrosine kinase Axl. Thus, induction of Rarres1 expression in endothelial cells is a prevalent molecular mechanism in human glomerulopathies and this seems to have a pathogenic role in driving inflammation and fibrosis via the Nuclear Factor-κB signaling pathway.
Collapse
Affiliation(s)
- Katja Möller-Hackbarth
- KI/AZ Integrated Cardio Metabolic Centre, Karolinska Institutet at Karolinska University Hospital, Huddinge, Stockholm, Sweden; Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet at Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Dina Dabaghie
- KI/AZ Integrated Cardio Metabolic Centre, Karolinska Institutet at Karolinska University Hospital, Huddinge, Stockholm, Sweden; Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet at Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Emmanuelle Charrin
- KI/AZ Integrated Cardio Metabolic Centre, Karolinska Institutet at Karolinska University Hospital, Huddinge, Stockholm, Sweden; Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet at Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Sonia Zambrano
- KI/AZ Integrated Cardio Metabolic Centre, Karolinska Institutet at Karolinska University Hospital, Huddinge, Stockholm, Sweden; Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet at Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Guillem Genové
- KI/AZ Integrated Cardio Metabolic Centre, Karolinska Institutet at Karolinska University Hospital, Huddinge, Stockholm, Sweden; Department of Medicine Huddinge, Division of Pathology, Karolinska Institutet at Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Xidan Li
- KI/AZ Integrated Cardio Metabolic Centre, Karolinska Institutet at Karolinska University Hospital, Huddinge, Stockholm, Sweden; Department of Medicine Huddinge, Division of Pathology, Karolinska Institutet at Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Annika Wernerson
- Department of Clinical Sciences, Division of Renal Medicine, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Mark Lal
- Bioscience Renal, Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), R&D Biopharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - Jaakko Patrakka
- KI/AZ Integrated Cardio Metabolic Centre, Karolinska Institutet at Karolinska University Hospital, Huddinge, Stockholm, Sweden; Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet at Karolinska University Hospital, Huddinge, Stockholm, Sweden.
| |
Collapse
|
19
|
Whitehead AJ, Engler AJ. Regenerative cross talk between cardiac cells and macrophages. Am J Physiol Heart Circ Physiol 2021; 320:H2211-H2221. [PMID: 33769920 DOI: 10.1152/ajpheart.00056.2021] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Aside from the first week postnatal, murine heart regeneration is restricted and responses to damage follow classic fibrotic remodeling. Recent transcriptomic analyses have suggested that significant cross talk with the sterile immune response could maintain a more embryonic-like signaling network that promotes acute, transient responses. However, with age, this response-likely mediated by neonatal yolk sac macrophages-then transitions to classical macrophage-mediated, cardiac fibroblast (CF)-based remodeling of the extracellular matrix (ECM) after myocardial infarction (MI). The molecular mechanisms that govern the change with age and drive fibrosis via inflammation are poorly understood. Using multiple ribonucleic acid sequencing (RNA-Seq) datasets, we attempt to resolve the relative contributions of CFs and macrophages in the bulk-healing response of regenerative (postnatal day 1) and nonregenerative hearts (postnatal day 8+). We performed an analysis of bulk RNA-Seq datasets from myocardium and cardiac fibroblasts as well as a single-cell RNA-Seq dataset from cardiac macrophages. MI-specific pathway differences revealed that nonregenerative hearts generated more ECM and had larger matricellular responses correlating with inflammation, produced greater chemotactic gradients to recruit macrophages, and expressed receptors for danger-associated molecular patterns at higher levels than neonates. These changes could result in elevated stress-response pathways compared with neonates, converging at NF-κB and activator protein-1 (AP-1) signaling. Profibrotic gene programs, which greatly diverge on day 3 post MI, lay the foundation for chronic fibrosis, and thus postnatal hearts older than 7 days typically exhibit significantly less regeneration. Our analyses suggest that the macrophage ontogenetic shift in the heart postnatally could result in detrimental stress signaling that suppresses regeneration.NEW & NOTEWORTHY Immediately postnatal mammalian hearts are able to regenerate after infarction, but the cells, pathways, and molecules that regulate this behavior are unclear. By comparing RNA-Seq datasets from regenerative mouse hearts and older, nonregenerative hearts, we are able to identify biological processes that are hallmarks of regeneration. We find that sterile inflammatory processes are upregulated in nonregenerative hearts, initiating profibrotic gene programs 3 days after myocardial infarction that can cause myocardial disease.
Collapse
Affiliation(s)
- Alexander J Whitehead
- Department of Bioengineering, University of California, San Diego, La Jolla, California.,Sanford Consortium for Regenerative Medicine, La Jolla, California
| | - Adam J Engler
- Department of Bioengineering, University of California, San Diego, La Jolla, California.,Sanford Consortium for Regenerative Medicine, La Jolla, California
| |
Collapse
|
20
|
Abed A, Leroyer AS, Kavvadas P, Authier F, Bachelier R, Foucault-Bertaud A, Bardin N, Cohen CD, Lindenmeyer MT, Genest M, Joshkon A, Jourde-Chiche N, Burtey S, Blot-Chabaud M, Dignat-George F, Chadjichristos CE. Endothelial-Specific Deletion of CD146 Protects Against Experimental Glomerulonephritis in Mice. Hypertension 2021; 77:1260-1272. [PMID: 33689459 DOI: 10.1161/hypertensionaha.119.14176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Ahmed Abed
- From the INSERM UMR-S1155, Tenon Hospital, Paris, France (A.A., P.K., F.A., M.G., C.E.C.).,Sorbonne Université, Paris, France (A.A., C.E.C.)
| | - Aurélie S Leroyer
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.)
| | - Panagiotis Kavvadas
- From the INSERM UMR-S1155, Tenon Hospital, Paris, France (A.A., P.K., F.A., M.G., C.E.C.)
| | - Florence Authier
- From the INSERM UMR-S1155, Tenon Hospital, Paris, France (A.A., P.K., F.A., M.G., C.E.C.)
| | - Richard Bachelier
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.)
| | - Alexandrine Foucault-Bertaud
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.)
| | - Nathalie Bardin
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.)
| | - Clemens D Cohen
- Nephrological Center, Medical Clinic and Policlinic IV, University of Munich, Germany (C.D.C.)
| | - Maja T Lindenmeyer
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Germany (M.T.L.)
| | - Magali Genest
- From the INSERM UMR-S1155, Tenon Hospital, Paris, France (A.A., P.K., F.A., M.G., C.E.C.)
| | - Ahmad Joshkon
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.)
| | - Noémie Jourde-Chiche
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.).,Department of Nephrology, Aix-Marseille University, AP-HM Hôpital de la Conception, Marseille, France (N.J.-C., S.B.)
| | - Stéphane Burtey
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.).,Department of Nephrology, Aix-Marseille University, AP-HM Hôpital de la Conception, Marseille, France (N.J.-C., S.B.)
| | - Marcel Blot-Chabaud
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.)
| | - Françoise Dignat-George
- Aix-Marseille University, INSERM 1263, INRAE 1260, C2VN, France (A.S.L., R.B., A.F.-B., N.B., A.J., N.J.-C., S.B., M.B.-C., F.D.-G.)
| | - Christos E Chadjichristos
- From the INSERM UMR-S1155, Tenon Hospital, Paris, France (A.A., P.K., F.A., M.G., C.E.C.).,Sorbonne Université, Paris, France (A.A., C.E.C.)
| |
Collapse
|
21
|
Ma WQ, Sun XJ, Zhu Y, Liu NF. Metformin attenuates hyperlipidaemia-associated vascular calcification through anti-ferroptotic effects. Free Radic Biol Med 2021; 165:229-242. [PMID: 33513420 DOI: 10.1016/j.freeradbiomed.2021.01.033] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 01/13/2021] [Accepted: 01/17/2021] [Indexed: 12/14/2022]
Abstract
Ferroptosis is a form of regulated cell death that involves metabolic dysfunction resulting from iron-dependent excessive lipid peroxidation. Elevated plasma levels of free fatty acids are tightly associated with cardiometabolic risk factors in patients with obesity, diabetes mellitus, and metabolic syndrome. Metformin (Met) is an antidiabetic drug with beneficial cardiovascular disease effects. The aim of this study was to determine the effects of Met on ferroptosis induced by lipid overload and the effects of these changes on vascular smooth muscle cells (VSMCs) calcification. We developed a hyperlipidaemia-related vascular calcification in vivo model with rats fed a high-fat diet combined with vitamin D3 plus nicotine, and palmitic acid (PA), the most abundant long-chain saturated fatty acid in plasma, was used to induce lipid overload and develop an oxidative stress-related calcification model in vitro. The results showed that Met inhibits hyperlipidaemia-associated calcium deposition in the rat aortic tissue. In vitro, treatment of VSMCs with PA stimulates ferroptosis concomitant with increased calcium deposition in VSMCs, while pretreatment with Met attenuates these effects. Furthermore, PA also promotes the protein expression of the extracellular matrix protein periostin (POSTN) and its secretion into the extracellular environment. More importantly, upregulation of POSTN increased the sensitivity of cells to ferroptosis. Mechanistically, upregulation of POSTN suppresses SLC7A11 expression through the inhibition of p53 in VSMCs, which contributes to a decrease in glutathione synthesis and therefore triggers ferroptosis. Interestingly, overexpression of p53 attenuates the inhibitory effect of POSTN on SLC7A11 expression, accompanied by increased Gpx4 expression. Furthermore, p53 knockdown suppresses Met-mediated anti-ferroptosis effects in PA-treated VSMCs, which may be related to the downregulation of SLC7A11 expression. In addition, supplementation of VSMCs with Met enhances the antioxidative capacity of VSMCs through Nrf2 signalling activation. Collectively, targeting POSTN in VSMCs may provide a new strategy for vascular calcification prevention or treatment.
Collapse
Affiliation(s)
- Wen-Qi Ma
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University 87 Dingjiaqiao, Nanjing, 210009, PR China
| | - Xue-Jiao Sun
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University 87 Dingjiaqiao, Nanjing, 210009, PR China
| | - Yi Zhu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University 87 Dingjiaqiao, Nanjing, 210009, PR China
| | - Nai-Feng Liu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University 87 Dingjiaqiao, Nanjing, 210009, PR China.
| |
Collapse
|
22
|
Arslan R, Karsiyaka Hendek M, Kisa U, Olgun E. The effect of non-surgical periodontal treatment on gingival crevicular fluid periostin levels in patients with gingivitis and periodontitis. Oral Dis 2020; 27:1478-1486. [PMID: 33012041 DOI: 10.1111/odi.13664] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 09/14/2020] [Accepted: 09/17/2020] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The objective of the study was to evaluate the effect of non-surgical periodontal treatment on gingival crevicular fluid (GCF) periostin levels in patients with gingivitis (G) and periodontitis (P). SUBJECTS AND METHODS A total of 90 subjects, 30 patients with P, 30 with G, and 30 periodontally healthy (H) subjects were included. Patients with periodontal disease received non-surgical periodontal treatment. GCF periostin levels were assessed at baseline, at the 6th week, and the 3rd month after treatment. RESULTS It was found that GCF periostin level was the lowest in the H group (89.31[47.12] pg/30 sec), followed by the G group (132.82[145.14] pg/30 sec), and the highest in the P group (207.75[189.45] pg/30 sec). These differences were statistically significant between H and the other groups (p < .001). After treatment, GCF periostin levels significantly decreased at the 6th week and the 3rd month in the G group, at the 3rd month in the P group compared to baseline values (p < .05). CONCLUSION The results of this study suggest that GCF periostin plays a role as a reliable biological marker in the pathogenesis of periodontal disease and non-surgical periodontal treatment is effective in decreasing GCF periostin levels.
Collapse
Affiliation(s)
- Rana Arslan
- Oral and Dental Health Center, Yozgat, Turkey
| | | | - Ucler Kisa
- Department of Biochemistry, Faculty of Medicine, Kirikkale University, Kirikkale, Turkey
| | - Ebru Olgun
- Department of Periodontology, Faculty of Dentistry, Kirikkale University, Kirikkale, Turkey
| |
Collapse
|
23
|
Sunadome H, Matsumoto H, Tachikawa R, Matsumoto T, Tanizawa K, Oga T, Ono J, Ohta S, Izuhara K, Hirai T, Chin K. Role of serum periostin in severe obstructive sleep apnea with albuminuria: an observational study. Respir Res 2020; 21:143. [PMID: 32517742 PMCID: PMC7285606 DOI: 10.1186/s12931-020-01413-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/03/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Periostin is a matricellular protein and is a useful marker in respiratory diseases. However, the roles of periostin in patients with obstructive sleep apnea (OSA) remain unclear. Several in vitro studies have suggested that mechanical stress, hypoxia, impaired metabolism, and kidney injury, which often accompany OSA, may upregulate the expression of periostin. Meanwhile, serum periostin level has been negatively associated with body mass index (BMI) in the general population. In this study, we hypothesized that a high level of serum periostin despite being overweight/obese may discriminate severe OSA or OSA with comorbidities from mild OSA with obesity alone. We aimed to clarify the roles of periostin in patients with OSA to assist in elucidating the heterogeneity of OSA with comorbidities. METHODS Among patients diagnosed as OSA, we examined the associations between serum periostin levels and clinical indices, including the severity of OSA, BMI, and comorbidities, using a multifaceted approach. The serum periostin levels and clinical indices were assessed after 3 months of continuous positive airway pressure (CPAP) treatment. RESULTS In 96 patients with OSA, serum periostin level was negatively correlated with BMI, albeit marginally, and tended to be higher in severe OSA than in others when adjusted for BMI. Cluster analysis identified four clusters, including two severe OSA clusters, one of which was characterized by high serum periostin levels and the presence of comorbidities, including albuminuria. In a comparative analysis of severe OSA cases (n = 53), the level of serum-free fatty acids and the frequency of albuminuria were higher in patients with high serum periostin level of ≥87 ng/mL, which was the highest quintile among all participants, than in those with low serum periostin levels (< 87 ng/mL, n = 41). In patients with severe OSA and high serum periostin levels, the levels of serum periostin and urinary albumin significantly decreased after 3 months of CPAP treatment. CONCLUSIONS Elevated serum periostin in patients with OSA despite being overweight/obese may be an indicator of severe OSA with comorbidities, particularly albuminuria.
Collapse
Affiliation(s)
- Hironobu Sunadome
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto city, Kyoto prefecture, 606-8507, Japan
| | - Hisako Matsumoto
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto city, Kyoto prefecture, 606-8507, Japan.
| | - Ryo Tachikawa
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto city, Kyoto prefecture, 606-8507, Japan.,Department of Respiratory Care and Sleep Control Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto City, Kyoto prefecture, 606-8507, Japan
| | - Takeshi Matsumoto
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto city, Kyoto prefecture, 606-8507, Japan.,Department of Respiratory Care and Sleep Control Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto City, Kyoto prefecture, 606-8507, Japan
| | - Kiminobu Tanizawa
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto city, Kyoto prefecture, 606-8507, Japan.,Department of Respiratory Care and Sleep Control Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto City, Kyoto prefecture, 606-8507, Japan
| | - Toru Oga
- Department of Respiratory Care and Sleep Control Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto City, Kyoto prefecture, 606-8507, Japan
| | - Junya Ono
- Shino-Test Corporation, 2-29-4 Oonodai, Minami-ku, Sagamihara City, Kanagawa prefecture, 252-0331, Japan
| | - Shoichiro Ohta
- Department of Laboratory Medicine, Saga Medical School, 5-1-1 Nabeshima, Saga City, Saga prefecture, 840-8502, Japan
| | - Kenji Izuhara
- Division of Biochemistry, Department of Biomolecular Science, Saga Medical School, 5-1-1 Nabeshima, Saga City, Saga prefecture, 840-8502, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto city, Kyoto prefecture, 606-8507, Japan
| | - Kazuo Chin
- Department of Respiratory Care and Sleep Control Medicine, Graduate School of Medicine, Kyoto University, 54 Kawaharacho, Shogoin, Sakyo-ku, Kyoto City, Kyoto prefecture, 606-8507, Japan
| |
Collapse
|
24
|
Jia Y, Gao L, Yang X, Zhang F, Chen A, Wang S, Shao J, Tan S, Zheng S. Blockade of periostin-dependent migration and adhesion by curcumol via inhibition of nuclear factor kappa B signaling in hepatic stellate cells. Toxicology 2020; 440:152475. [PMID: 32344006 DOI: 10.1016/j.tox.2020.152475] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/18/2020] [Accepted: 04/20/2020] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Curcumol, a guaiane-type sesquiterpenoid hemiketal extracted from the herb Rhizoma Curcumae, exhibits multiple-pharmacological activities. We previously reported that curcumol ameliorated hepatic fibrosis by inhibiting hepatic stellate cell (HSC) activation. In this study, we aimed to investigate the effect of curcumol on HSC migration and adhesion, and reveal its regulation mechanisms. MATERIALS AND METHODS Cellular viability was determined by Cell Counting Kit-8. Cell migration was detected by boyden chamber and cell scratch experiment. Recombinant human periostin (rh POSTN) and adeno-associated viral (AAV)-GFP-periostin were used to achieve POSTN overexpression in vitro and in vivo, respectively. Nuclear factor kappa B (NF-κB)-p65 overexpression was achieved by using plasmid. ELISA was conducted to detect POSTN level. Immunohistochemistry, qRT-PCR, Western blotting, and immunofluorescence were performed to assess associated factor expression. RESULTS Curcumol suppressed HSC migration and adhesion, and reduced the secretion and expression of POSTN. By gain of function POSTN in HSCs, using rh POSTN, we found that the inhibition of HSC migration and adhesion by curcumol depended on the decrease of POSTN. Besides, curcumol protection against chronic CCl4-caused hepatic fibrosis could be impaired by POSTN overexpression. Moreover, we showed that curcumol repressed NF-κB signaling and the production of pro-inflammatory factor. Importantly, curcumol down-regulation of POSTN was rescued by knock-in of NF-κB, as well as the inhibition of HSC migration and adhesion. CONCLUSION These findings reveal the molecular mechanism of curcumol-reduced HSC migration and adhesion, by which points to the possibility of using curcumol based on NF-κB dependent POSTN for the treatment of fibrogenesis.
Collapse
Affiliation(s)
- Yan Jia
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Liyuan Gao
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Xiang Yang
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Feng Zhang
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China; Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, China; Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, China
| | - Anping Chen
- Department of Pathology, School of Medicine, Saint Louis University, St Louis, MO 63104, USA
| | - Shijun Wang
- Shandong Co-Innovation Center of TCM Formula, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, China
| | - Jiangjuan Shao
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China.
| | - Shanzhong Tan
- Department of Hepatology, Integrated Traditional Chinese and Western Medicine, Nanjing Second Hospital, China.
| | - Shizhong Zheng
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China; Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, China; Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, China.
| |
Collapse
|
25
|
Guo K, Pan P, Wu M, Ma Y, Lu J, Chen H. Hyposialylated angiopoietin-like-4 induces apoptosis of podocytes via β1 Integrin/FAK signaling in diabetic nephropathy. Mol Cell Endocrinol 2020; 505:110730. [PMID: 31981598 DOI: 10.1016/j.mce.2020.110730] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 01/18/2020] [Accepted: 01/20/2020] [Indexed: 12/12/2022]
Abstract
Angiopoietin-like-4 (ANGPTL4) is reported to mediate proteinuria in some types of glomerulonephropathy. However, the mechanism underlying the effect on podocytes of ANGPTL4 under pathologic conditions in diabetic nephropathy (DN) is unclear. We investigated the role of ANGPTL4 in the pathogenesis of DN. In DN rats, elevated ANGPTL4 expression was associated with increased proteinuria, glomerular hypertrophy, and ultrastructural changes in podocytes. In vitro, hyperglycemia induced the upregulation of ANGPTL4, which led to activation of integrin-β1/FAK signaling with increased apoptosis of podocytes and actin cytoskeleton derangement. These pathological changes were reversed by transfection with a lentivirus expressing short hairpin RNA against integrin-β1 or an ANGPTL4-neutralizing antibody in vitro. Furthermore, supplementation with the sialic acid precursor ManNAc reversed these pathological changes and conferred renoprotection in a mouse model of DN. Our findings suggest that ANGPTL4 mediates high glucose-induced loss of podocytes by modulating their detachment and apoptosis in vivo and in vitro. This study deepens our understanding of the mechanisms of podocyte loss in DN and shows targeting ANGPTL4-related signaling has therapeutic potential for DN.
Collapse
Affiliation(s)
- Kaifeng Guo
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Department of Endocrinology and Metabolism, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, 200233, China; Department of Endocrinology and Metabolism, Minhang Hospital, Fudan University; Minhang Branch, Zhongshan Hospital, Fudan University; Central Hospital of Minhang District, Shanghai, 201199, China
| | - Pan Pan
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Department of Endocrinology and Metabolism, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Mian Wu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Department of Endocrinology and Metabolism, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Yiwen Ma
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Department of Endocrinology and Metabolism, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Junxi Lu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Department of Endocrinology and Metabolism, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Haibing Chen
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Department of Endocrinology and Metabolism, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| |
Collapse
|
26
|
Li L, Wang R, Jia Y, Rong R, Xu M, Zhu T. Exosomes Derived From Mesenchymal Stem Cells Ameliorate Renal Ischemic-Reperfusion Injury Through Inhibiting Inflammation and Cell Apoptosis. Front Med (Lausanne) 2019; 6:269. [PMID: 31867333 PMCID: PMC6907421 DOI: 10.3389/fmed.2019.00269] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 11/01/2019] [Indexed: 01/15/2023] Open
Abstract
This study aimed to investigate the underlying mechanism of mesenchymal stem cells (MSCs) on protection of renal ischemia reperfusion injury (IRI). Exosomes originated from MSCs (MSC-ex) were extracted according to the instructions of Total Exosome Isolation Reagent. Rats were divided into five groups: sham-operated, IRI, MSC, MSC-ex, and MSC-ex + RNAase group. MSCs or MSC-ex were injected via carotid artery. The renal function test and pathological detection were applied to determine the renoprotection of MSC-ex on IRI. Western blotting and quantitative reverse transcription polymerase chain reaction (RT-qPCR) were conducted to examine the levels of apoptosis-related proteins and inflammatory cytokines. Our results revealed that MSC-derived exosomes attenuated renal dysfunction, histologic damage, and decreased apoptosis. The expression levels of inflammatory cytokines, such as interleukin 6 (IL-6), tumor necrosis factor-α (TNF-α), nuclear factor kappa B (NF-κB), and interferon gamma (IFN-γ), were decreased by the MSC-ex treatment. The expression levels of caspase-9, cleaved caspase-3, Bax, and Bcl-2 caused by IR were also inhibited by MSC-ex. MSC-ex + RNAase group shared the similar pattern of changes with IRI group, likely due to the ability of RNA hydrolase to eliminate the function of exosomes. Our results demonstrated that exosomes originating from MSCs have protective effects on IRI via inhibiting cell apoptosis and inflammatory responses. Out findings may provide a new insight into therapeutic mechanism of MSCs on renal IRI.
Collapse
Affiliation(s)
- Long Li
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Rulin Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yichen Jia
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Ruiming Rong
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Ming Xu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Tongyu Zhu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| |
Collapse
|
27
|
Paunas FTI, Finne K, Leh S, Osman TAH, Marti HP, Berven F, Vikse BE. Characterization of glomerular extracellular matrix in IgA nephropathy by proteomic analysis of laser-captured microdissected glomeruli. BMC Nephrol 2019; 20:410. [PMID: 31726998 PMCID: PMC6854890 DOI: 10.1186/s12882-019-1598-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 10/23/2019] [Indexed: 12/29/2022] Open
Abstract
Background IgA nephropathy (IgAN) involves mesangial matrix expansion, but the proteomic composition of this matrix is unknown. The present study aimed to characterize changes in extracellular matrix in IgAN. Methods In the present study we used mass spectrometry-based proteomics in order to quantitatively compare protein abundance between glomeruli of patients with IgAN (n = 25) and controls with normal biopsy findings (n = 15). Results Using a previously published paper by Lennon et al. and cross-referencing with the Matrisome database we identified 179 extracellular matrix proteins. In the comparison between IgAN and controls, IgAN glomeruli showed significantly higher abundance of extracellular matrix structural proteins (e.g periostin, vitronectin, and extracellular matrix protein 1) and extracellular matrix associated proteins (e.g. azurocidin, myeloperoxidase, neutrophil elastase, matrix metalloproteinase-9 and matrix metalloproteinase 2). Periostin (fold change 3.3) and azurocidin (3.0) had the strongest fold change between IgAN and controls; periostin was also higher in IgAN patients who progressed to ESRD as compared to patients who did not. Conclusion IgAN is associated with widespread changes of the glomerular extracellular matrix proteome. Proteins important in glomerular sclerosis or inflammation seem to be most strongly increased and periostin might be an important marker of glomerular damage in IgAN.
Collapse
Affiliation(s)
- Flavia Teodora Ioana Paunas
- Department of Medicine, Haugesund Hospital, Haugesund, Norway. .,Department of Clinical Medicine, University of Bergen, Bergen, Norway.
| | - Kenneth Finne
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Sabine Leh
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | | | - Hans-Peter Marti
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Frode Berven
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Bjørn Egil Vikse
- Department of Medicine, Haugesund Hospital, Haugesund, Norway.,Department of Clinical Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
28
|
Wan J, Shan Y, Song X, Chen S, Lu X, Jin J, Su Q, Liu B, Sun W, Li B. Adipocyte-derived Periostin mediates glucocorticoid-induced hepatosteatosis in mice. Mol Metab 2019; 31:24-35. [PMID: 31918919 PMCID: PMC6880106 DOI: 10.1016/j.molmet.2019.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/21/2019] [Accepted: 11/01/2019] [Indexed: 02/06/2023] Open
Abstract
Objective Long-term glucocorticoids (GCs) therapy usually causes many metabolic side effects, including fatty liver. However, the molecular mechanisms remain poorly understood. Herein, we explored the molecular basis of GCs in the development of fatty liver. Methods C57BL/6 male mice were injected with Dexamethasone (DEX) while mouse primary hepatocytes (MPHs), HepG2 and Hep1-6 cells were cultured in the presence of DEX. Genes expression in liver tissues and hepatocytes were assessed by quantitative real-time PCR and western blotting, respectively. To explore whether Periostin is involved in the development of GCs-induced fatty liver, wild-type and Periostin knockout mice were treated with DEX or vehicle control. Luciferase reporter and chromatin immunoprecipitation assays were used to determine the regulatory roles of GCs on Periostin expression. Results We show that treatment of dexamethasone (DEX), a synthetic analog of GCs, led to the accumulation of triglycerides in the livers of mice, but not in cultured hepatocytes, suggesting that GCs may promote liver steatosis through integrative organ crosstalk mediated by systemic factors. We further found that DEX upregulated the expression levels of Periostin in white adipose tissues, which in turn promoted liver steatosis. Administration of a Periostin-neutralizing antibody or genetic ablation of Periostin largely attenuated DEX-induced hepatic steatosis in mice. Conclusions Our findings provided a novel insight that GCs could promote liver steatosis through integrative organ crosstalk mediated by white fat-secreted Periostin. These results establish Periostin as an endocrine factor with therapeutic potential for the treatment of GCs-associated fatty liver. Dexamethasone (DEX) treatment led to triglycerides accumulation in the liver of mice, but not in cultured hepatocytes. DEX treatment upregulates Periostin in white adipose tissues, which in turn induces liver steatosis in mice. Genetic ablation or pharmacological inhibition of Periostin partially attenuated DEX -induced hepatic steatosis in mice.
Collapse
Affiliation(s)
- Jian Wan
- Department of Emergency and Critical Care Medicine, Shanghai Pudong New Area People's Hospital, Shanghai University of Medicine and Health Sciences, Shanghai 201299, China
| | - Yi Shan
- Department of Emergency and ICU, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Xi Song
- Department of Emergency and Critical Care Medicine, Shanghai Pudong New Area People's Hospital, Shanghai University of Medicine and Health Sciences, Shanghai 201299, China
| | - Song Chen
- Department of Emergency and Critical Care Medicine, Shanghai Pudong New Area People's Hospital, Shanghai University of Medicine and Health Sciences, Shanghai 201299, China
| | - Xinyuan Lu
- Department of Emergency and Critical Care Medicine, Shanghai Pudong New Area People's Hospital, Shanghai University of Medicine and Health Sciences, Shanghai 201299, China
| | - Jie Jin
- Department of Endocrinology, XinHua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Qing Su
- Department of Endocrinology, XinHua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Bin Liu
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Huangshi, Hubei 435003, China
| | - Wanju Sun
- Department of Emergency and Critical Care Medicine, Shanghai Pudong New Area People's Hospital, Shanghai University of Medicine and Health Sciences, Shanghai 201299, China.
| | - Bo Li
- Department of Endocrinology, XinHua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| |
Collapse
|
29
|
Kormann R, Kavvadas P, Placier S, Vandermeersch S, Dorison A, Dussaule JC, Chadjichristos CE, Prakoura N, Chatziantoniou C. Periostin Promotes Cell Proliferation and Macrophage Polarization to Drive Repair after AKI. J Am Soc Nephrol 2019; 31:85-100. [PMID: 31690575 DOI: 10.1681/asn.2019020113] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 09/28/2019] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The matricellular protein periostin has been associated with CKD progression in animal models and human biopsy specimens. Periostin functions by interacting with extracellular matrix components to drive collagen fibrillogenesis and remodeling or by signaling through cell-surface integrin receptors to promote cell adhesion, migration, and proliferation. However, its role in AKI is unknown. METHODS We used mice with conditional tubule-specific overexpression of periostin or knockout mice lacking periostin expression in the renal ischemia-reperfusion injury model, and primary cultures of isolated tubular cells in a hypoxia-reoxygenation model. RESULTS Tubular epithelial cells showed strong production of periostin during the repair phase of ischemia reperfusion. Periostin overexpression protected mice from renal injury compared with controls, whereas knockout mice showed increased tubular injury and deteriorated renal function. Periostin interacted with its receptor, integrin-β1, to inhibit tubular cell cycle arrest and apoptosis in in vivo and in vitro models. After ischemia-reperfusion injury, periostin-overexpressing mice exhibited diminished expression of proinflammatory molecules and had more F4/80+ macrophages compared with knockout mice. Macrophages from periostin-overexpressing mice showed increased proliferation and expression of proregenerative factors after ischemia-reperfusion injury, whereas knockout mice exhibited the opposite. Coculturing a macrophage cell line with hypoxia-treated primary tubules overexpressing periostin, or treating such macrophages with recombinant periostin, directly induced macrophage proliferation and expression of proregenerative molecules. CONCLUSIONS In contrast to the detrimental role of periostin in CKD, we discovered a protective role of periostin in AKI. Our findings suggest periostin may be a novel and important mediator of mechanisms controlling renal repair after AKI.
Collapse
Affiliation(s)
- Raphaёl Kormann
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche Scientifique 1155, Tenon Hospital, Paris, France; and.,Faculty of Medicine, Sorbonne University, Paris, France
| | - Panagiotis Kavvadas
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche Scientifique 1155, Tenon Hospital, Paris, France; and
| | - Sandrine Placier
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche Scientifique 1155, Tenon Hospital, Paris, France; and
| | - Sophie Vandermeersch
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche Scientifique 1155, Tenon Hospital, Paris, France; and
| | - Aude Dorison
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche Scientifique 1155, Tenon Hospital, Paris, France; and.,Faculty of Medicine, Sorbonne University, Paris, France
| | - Jean-Claude Dussaule
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche Scientifique 1155, Tenon Hospital, Paris, France; and.,Faculty of Medicine, Sorbonne University, Paris, France
| | - Christos E Chadjichristos
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche Scientifique 1155, Tenon Hospital, Paris, France; and.,Faculty of Medicine, Sorbonne University, Paris, France
| | - Niki Prakoura
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche Scientifique 1155, Tenon Hospital, Paris, France; and
| | - Christos Chatziantoniou
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche Scientifique 1155, Tenon Hospital, Paris, France; and .,Faculty of Medicine, Sorbonne University, Paris, France
| |
Collapse
|
30
|
Prakoura N, Hadchouel J, Chatziantoniou C. Novel Targets for Therapy of Renal Fibrosis. J Histochem Cytochem 2019; 67:701-715. [PMID: 31116064 PMCID: PMC6713972 DOI: 10.1369/0022155419849386] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 04/15/2019] [Indexed: 12/19/2022] Open
Abstract
Renal fibrosis is an important component of chronic kidney disease, an incurable pathology with increasing prevalence worldwide. With a lack of available therapeutic options, end-stage renal disease is currently treated with renal replacement therapy through dialysis or transplantation. In recent years, many efforts have been made to identify novel targets for therapy of renal diseases, with special focus on the characterization of unknown mediators and pathways participating in renal fibrosis development. Using experimental models of renal disease and patient biopsies, we identified four novel mediators of renal fibrosis with potential to constitute future therapeutic targets against kidney disease: discoidin domain receptor 1, periostin, connexin 43, and cannabinoid receptor 1. The four candidates were highly upregulated in different models of renal disease and were localized at the sites of injury. Subsequent studies showed that they are centrally involved in the underlying mechanisms of renal fibrosis progression. Interestingly, inhibition of either of these proteins by different strategies, including gene deletion, antisense administration, or specific blockers, delayed the progression of renal disease and preserved renal structure and function, even when the inhibition started after initiation of the disease. This review will summarize the current findings on these candidates emphasizing on their potential to constitute future targets of therapy.
Collapse
Affiliation(s)
- Niki Prakoura
- Institut National de la Santé Et de la Recherche Médicale (INSERM) UMRS 1155, Tenon Hospital, Paris, France
| | - Juliette Hadchouel
- Institut National de la Santé Et de la Recherche Médicale (INSERM) UMRS 1155, Tenon Hospital, Paris, France
- Sorbonne Université, Paris, France
| | - Christos Chatziantoniou
- Institut National de la Santé Et de la Recherche Médicale (INSERM) UMRS 1155, Tenon Hospital, Paris, France
- Sorbonne Université, Paris, France
| |
Collapse
|
31
|
Lazareth H, Henique C, Lenoir O, Puelles VG, Flamant M, Bollée G, Fligny C, Camus M, Guyonnet L, Millien C, Gaillard F, Chipont A, Robin B, Fabrega S, Dhaun N, Camerer E, Kretz O, Grahammer F, Braun F, Huber TB, Nochy D, Mandet C, Bruneval P, Mesnard L, Thervet E, Karras A, Le Naour F, Rubinstein E, Boucheix C, Alexandrou A, Moeller MJ, Bouzigues C, Tharaux PL. The tetraspanin CD9 controls migration and proliferation of parietal epithelial cells and glomerular disease progression. Nat Commun 2019; 10:3303. [PMID: 31341160 PMCID: PMC6656772 DOI: 10.1038/s41467-019-11013-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 06/07/2019] [Indexed: 01/02/2023] Open
Abstract
The mechanisms driving the development of extracapillary lesions in focal segmental glomerulosclerosis (FSGS) and crescentic glomerulonephritis (CGN) remain poorly understood. A key question is how parietal epithelial cells (PECs) invade glomerular capillaries, thereby promoting injury and kidney failure. Here we show that expression of the tetraspanin CD9 increases markedly in PECs in mouse models of CGN and FSGS, and in kidneys from individuals diagnosed with these diseases. Cd9 gene targeting in PECs prevents glomerular damage in CGN and FSGS mouse models. Mechanistically, CD9 deficiency prevents the oriented migration of PECs into the glomerular tuft and their acquisition of CD44 and β1 integrin expression. These findings highlight a critical role for de novo expression of CD9 as a common pathogenic switch driving the PEC phenotype in CGN and FSGS, while offering a potential therapeutic avenue to treat these conditions. In both focal segmental glomerulosclerosis (FSGS) and crescentic glomerulonephritis (CGN), kidney injury is characterised by the invasion of glomerular tufts by parietal epithelial cells (PECs). Here Lazareth et al. identify the tetraspanin CD9 as a key regulator of PEC migration, and find its upregulation in FSGS and CGN contributes to kidney injury in both diseases.
Collapse
Affiliation(s)
- Hélène Lazareth
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France.,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France.,Renal Division, Georges Pompidou European Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris, Paris, F-75015, France.,Laboratoire d'Optique et Biosciences, Ecole polytechnique, CNRS UMR7645, INSERM U1182, Université Paris-Saclay, Palaiseau, F-91128, France
| | - Carole Henique
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France. .,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France. .,Institut Mondor de Recherche Biomédicale, Inserm U955, Equipe 21, Université Paris Est Créteil, Créteil, F-94010, France.
| | - Olivia Lenoir
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France.,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France
| | - Victor G Puelles
- Department of Nephrology and Clinical Immunology, University Hospital RWTH Aachen, Pauwelsstrasse 30, D-52074, Aachen, Germany.,Department of Medicine III, Faculty of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, D-20246, Germany.,Department of Nephrology and Center for Inflammatory Diseases, Monash University, Melbourne, VIC 3168, Australia
| | - Martin Flamant
- Xavier Bichat University Hospital, Université de Paris, Paris, F-75018, France
| | - Guillaume Bollée
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France.,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France
| | - Cécile Fligny
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France.,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France
| | - Marine Camus
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France.,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France
| | - Lea Guyonnet
- National Cytometry Platform, Department of Infection and Immunity, Luxembourg Institute of Health, Luxembourg, L-4354, Luxembourg
| | - Corinne Millien
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France.,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France
| | - François Gaillard
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France.,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France
| | - Anna Chipont
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France.,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France
| | - Blaise Robin
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France.,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France
| | - Sylvie Fabrega
- Université de Paris, Institut Imagine, Plateforme Vecteurs Viraux et Transfert de Gènes, IFR94, Hôpital Necker Enfants-Malades, Paris, F-75015, France
| | - Neeraj Dhaun
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, EH16 4SA, Scotland, UK
| | - Eric Camerer
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France.,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France
| | - Oliver Kretz
- Department of Medicine III, Faculty of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, D-20246, Germany.,Renal Division, Faculty of Medicine, Medical Centre, University of Freiburg, Freiburg, D-79106, Germany
| | - Florian Grahammer
- Department of Medicine III, Faculty of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, D-20246, Germany.,Renal Division, Faculty of Medicine, Medical Centre, University of Freiburg, Freiburg, D-79106, Germany
| | - Fabian Braun
- Department of Medicine III, Faculty of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, D-20246, Germany.,Renal Division, Faculty of Medicine, Medical Centre, University of Freiburg, Freiburg, D-79106, Germany
| | - Tobias B Huber
- Department of Medicine III, Faculty of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, D-20246, Germany.,Renal Division, Faculty of Medicine, Medical Centre, University of Freiburg, Freiburg, D-79106, Germany
| | - Dominique Nochy
- Department of Pathology, Georges Pompidou European Hospital, Assistance Publique-Hôpitaux de Paris, Paris, F-75015, France
| | - Chantal Mandet
- Department of Pathology, Georges Pompidou European Hospital, Assistance Publique-Hôpitaux de Paris, Paris, F-75015, France
| | - Patrick Bruneval
- Department of Pathology, Georges Pompidou European Hospital, Assistance Publique-Hôpitaux de Paris, Paris, F-75015, France
| | - Laurent Mesnard
- Critical Care Nephrology and Kidney Transplantation, Hôpital Tenon, Assistance Publique-Hôpitaux de Paris, Unité Mixte de Recherche S1155, Pierre and Marie Curie University, Paris, F-75020, France
| | - Eric Thervet
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France.,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France.,Renal Division, Georges Pompidou European Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris, Paris, F-75015, France
| | - Alexandre Karras
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France.,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France.,Renal Division, Georges Pompidou European Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris, Paris, F-75015, France
| | | | - Eric Rubinstein
- Inserm U935, Université Paris-Sud, Villejuif, F-94800, France
| | - Claude Boucheix
- Inserm U935, Université Paris-Sud, Villejuif, F-94800, France
| | - Antigoni Alexandrou
- Laboratoire d'Optique et Biosciences, Ecole polytechnique, CNRS UMR7645, INSERM U1182, Université Paris-Saclay, Palaiseau, F-91128, France
| | - Marcus J Moeller
- Department of Nephrology and Clinical Immunology, University Hospital RWTH Aachen, Pauwelsstrasse 30, D-52074, Aachen, Germany
| | - Cédric Bouzigues
- Laboratoire d'Optique et Biosciences, Ecole polytechnique, CNRS UMR7645, INSERM U1182, Université Paris-Saclay, Palaiseau, F-91128, France
| | - Pierre-Louis Tharaux
- Institut National de la Santé et de la Recherche Médicale (Inserm), Unit 970, Paris Cardiovascular Center - PARCC, 56 rue Leblanc, F-75015, Paris, France. .,Université de Paris, UMR-S970, 56 rue Leblanc, F-75015, Paris, France. .,Renal Division, Georges Pompidou European Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris, Paris, F-75015, France.
| |
Collapse
|
32
|
Bian X, Bai Y, Su X, Zhao G, Sun G, Li D. Knockdown of periostin attenuates 5/6 nephrectomy‐induced intrarenal renin–angiotensin system activation, fibrosis, and inflammation in rats. J Cell Physiol 2019; 234:22857-22873. [PMID: 31127625 DOI: 10.1002/jcp.28849] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 04/30/2019] [Accepted: 04/30/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Xiaohui Bian
- Department of Nephrology Shengjing Hospital of China Medical University Shenyang P.R. China
| | - Yu Bai
- Department of Nephrology Shengjing Hospital of China Medical University Shenyang P.R. China
| | - Xiaoxiao Su
- Department of Nephrology Shengjing Hospital of China Medical University Shenyang P.R. China
| | - Guifeng Zhao
- Research Center Shengjing Hospital of China Medical University Shenyang P.R. China
| | - Guangping Sun
- Department of Nephrology Shengjing Hospital of China Medical University Shenyang P.R. China
| | - Detian Li
- Department of Nephrology Shengjing Hospital of China Medical University Shenyang P.R. China
| |
Collapse
|
33
|
Bülow RD, Boor P. Extracellular Matrix in Kidney Fibrosis: More Than Just a Scaffold. J Histochem Cytochem 2019; 67:643-661. [PMID: 31116062 DOI: 10.1369/0022155419849388] [Citation(s) in RCA: 243] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Kidney fibrosis is the common histological end-point of progressive, chronic kidney diseases (CKDs) regardless of the underlying etiology. The hallmark of renal fibrosis, similar to all other organs, is pathological deposition of extracellular matrix (ECM). Renal ECM is a complex network of collagens, elastin, and several glycoproteins and proteoglycans forming basal membranes and interstitial space. Several ECM functions beyond providing a scaffold and organ stability are being increasingly recognized, for example, in inflammation. ECM composition is determined by the function of each of the histological compartments of the kidney, that is, glomeruli, tubulo-interstitium, and vessels. Renal ECM is a dynamic structure undergoing remodeling, particularly during fibrosis. From a clinical perspective, ECM proteins are directly involved in several rare renal diseases and indirectly in CKD progression during renal fibrosis. ECM proteins could serve as specific non-invasive biomarkers of fibrosis and scaffolds in regenerative medicine. The gold standard and currently only specific means to measure renal fibrosis is renal biopsy, but new diagnostic approaches are appearing. Here, we discuss the localization, function, and remodeling of major renal ECM components in healthy and diseased, fibrotic kidneys and the potential use of ECM in diagnostics of renal fibrosis and in tissue engineering.
Collapse
Affiliation(s)
- Roman David Bülow
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Peter Boor
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany.,Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
34
|
Bian X, Su X, Wang Y, Zhao G, Zhang B, Li D. Periostin contributes to renal and cardiac dysfunction in rats with chronic kidney disease: Reduction of PPARα. Biochimie 2019; 160:172-182. [DOI: 10.1016/j.biochi.2019.03.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 03/06/2019] [Indexed: 12/20/2022]
|
35
|
Abstract
The global burden of chronic kidney disease will increase during the next century. As NFκB, first described more than 30 years ago, plays a major role in immune and non-immune-mediated diseases and in inflammatory and metabolic disorders, this review article summarizes current knowledge on the role of NFκB in in vivo kidney injury and describes the new and so far not completely understood crosstalk between canonical and non-canonical NFκB pathways in T-lymphocyte activation in renal disease.
Collapse
Affiliation(s)
- Ning Song
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Friedrich Thaiss
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Linlin Guo
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
36
|
Allard DE, Wang Y, Li JJ, Conley B, Xu EW, Sailer D, Kimpston C, Notini R, Smith CJ, Koseoglu E, Starmer J, Zeng XL, Howard JF, Hoke A, Scherer SS, Su MA. Schwann cell-derived periostin promotes autoimmune peripheral polyneuropathy via macrophage recruitment. J Clin Invest 2018; 128:4727-4741. [PMID: 30222134 PMCID: PMC6159985 DOI: 10.1172/jci99308] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 07/31/2018] [Indexed: 12/12/2022] Open
Abstract
Chronic inflammatory demyelinating polyneuropathy (CIDP) and Guillain-Barre syndrome (GBS) are inflammatory neuropathies that affect humans and are characterized by peripheral nerve myelin destruction and macrophage-containing immune infiltrates. In contrast to the traditional view that the peripheral nerve is simply the target of autoimmunity, we report here that peripheral nerve Schwann cells exacerbate the autoimmune process through extracellular matrix (ECM) protein induction. In a spontaneous autoimmune peripheral polyneuropathy (SAPP) mouse model of inflammatory neuropathy and CIDP nerve biopsies, the ECM protein periostin (POSTN) was upregulated in affected sciatic nerves and was primarily expressed by Schwann cells. Postn deficiency delayed the onset and reduced the extent of neuropathy, as well as decreased the number of macrophages infiltrating the sciatic nerve. In an in vitro assay, POSTN promoted macrophage chemotaxis in an integrin-AM (ITGAM) and ITGAV-dependent manner. The PNS-infiltrating macrophages in SAPP-affected nerves were pathogenic, since depletion of macrophages protected against the development of neuropathy. Our findings show that Schwann cells promote macrophage infiltration by upregulating Postn and suggest that POSTN is a novel target for the treatment of macrophage-associated inflammatory neuropathies.
Collapse
Affiliation(s)
| | - Yan Wang
- Department of Pediatrics, University of North Carolina at Chapel Hill (UNC-CH), Chapel Hill, North Carolina, USA
| | - Jian Joel Li
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Bridget Conley
- Department of Pediatrics, University of North Carolina at Chapel Hill (UNC-CH), Chapel Hill, North Carolina, USA
| | - Erin W. Xu
- Department of Pediatrics, University of North Carolina at Chapel Hill (UNC-CH), Chapel Hill, North Carolina, USA
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, California, USA
| | - David Sailer
- Department of Pediatrics, University of North Carolina at Chapel Hill (UNC-CH), Chapel Hill, North Carolina, USA
| | - Caellaigh Kimpston
- Department of Pediatrics, University of North Carolina at Chapel Hill (UNC-CH), Chapel Hill, North Carolina, USA
| | - Rebecca Notini
- Department of Pediatrics, University of North Carolina at Chapel Hill (UNC-CH), Chapel Hill, North Carolina, USA
| | | | - Emel Koseoglu
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, USA
- Neurology Department, School of Medicine, Erciyes University, Kayseri, Turkey
| | | | - Xiaopei L. Zeng
- Department of Pediatrics, University of North Carolina at Chapel Hill (UNC-CH), Chapel Hill, North Carolina, USA
| | - James F. Howard
- Department of Neurology, UNC-CH, Chapel Hill, North Carolina, USA
| | - Ahmet Hoke
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Steven S. Scherer
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Maureen A. Su
- Department of Microbiology and Immunology and
- Department of Pediatrics, University of North Carolina at Chapel Hill (UNC-CH), Chapel Hill, North Carolina, USA
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, California, USA
- Department of Pediatrics, UCLA, Los Angeles, California, USA
| |
Collapse
|
37
|
Ni Y, Wang X, Yin X, Li Y, Liu X, Wang H, Liu X, Zhang J, Gao H, Shi B, Zhao S. Plectin protects podocytes from adriamycin-induced apoptosis and F-actin cytoskeletal disruption through the integrin α6β4/FAK/p38 MAPK pathway. J Cell Mol Med 2018; 22:5450-5467. [PMID: 30187999 PMCID: PMC6201223 DOI: 10.1111/jcmm.13816] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 01/16/2018] [Accepted: 06/29/2018] [Indexed: 02/06/2023] Open
Abstract
Podocyte injury is an early pathological change characteristic of various glomerular diseases, and apoptosis and F‐actin cytoskeletal disruption are typical features of podocyte injury. In this study, we found that adriamycin (ADR) treatment resulted in typical podocyte injury and repressed plectin expression. Restoring plectin expression protected against ADR‐induced podocyte injury whereas siRNA‐mediated plectin silencing produced similar effects as ADR‐induced podocyte injury, suggesting that plectin plays a key role in preventing podocyte injury. Further analysis showed that plectin repression induced significant integrin α6β4, focal adhesion kinase (FAK) and p38 MAPK phosphorylation. Mutating Y1494, a key tyrosine residue in the integrin β4 subunit, blocked FAK and p38 phosphorylation, thereby alleviating podocyte injury. Inhibitor studies demonstrated that FAK Y397 phosphorylation promoted p38 activation, resulting in podocyte apoptosis and F‐actin cytoskeletal disruption. In vivo studies showed that administration of ADR to rats resulted in significantly increased 24‐hour urine protein levels along with decreased plectin expression and activated integrin α6β4, FAK, and p38. Taken together, these findings indicated that plectin protects podocytes from ADR‐induced apoptosis and F‐actin cytoskeletal disruption by inhibiting integrin α6β4/FAK/p38 pathway activation and that plectin may be a therapeutic target for podocyte injury‐related glomerular diseases.
Collapse
Affiliation(s)
- Yongliang Ni
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China.,Department of Urology, Shandong Provincial Third Hospital, Jinan, Shandong, China
| | - Xin Wang
- Department of Urology, Tengzhou Central People's Hospital affiliated to Jining Medical College, Xintan Road 181, Tengzhou, China
| | - Xiaoxuan Yin
- Department of Traditional Chinese Medicine, Yankuang Group General Hospital, Zoucheng, China
| | - Yan Li
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Xigao Liu
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Haixin Wang
- Department of Urology, Yankuang Group General Hospital, Zoucheng, China
| | - Xiangjv Liu
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China.,Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University
| | - Jun Zhang
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China.,Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University
| | - Haiqing Gao
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China.,Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University
| | - Benkang Shi
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Shaohua Zhao
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong, China.,Key Laboratory of Cardiovascular Proteomics of Shandong Province, Qilu Hospital of Shandong University
| |
Collapse
|
38
|
Ding Y, Ge Q, Qu H, Feng Z, Long J, Wei Q, Zhou Q, Wu R, Yao L, Deng H. Increased serum periostin concentrations are associated with the presence of diabetic retinopathy in patients with type 2 diabetes mellitus. J Endocrinol Invest 2018; 41:937-945. [PMID: 29349642 DOI: 10.1007/s40618-017-0820-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 12/27/2017] [Indexed: 12/12/2022]
Abstract
PURPOSE To determine the association between serum periostin and the presence of diabetic retinopathy (DR). METHODS Serum periostin was detected in 114 healthy subjects, 122 patients with type 2 diabetes mellitus (T2DM) and 159 patients with DR and compared among groups. Clinical data and other laboratory measurements such as glycated hemoglobin (HbA1c), lipid profiles, serum creatinine (Cr) and high-sensitivity CRP (hsCRP) were also collected and compared among groups. For subgroup analysis, patients with DR were divided into a non-proliferated diabetic retinopathy (NPDR) group and a proliferated diabetic retinopathy (PDR) group. Multivariate analysis was performed using logistic regression models. RESULTS The serum periostin level was significantly higher in patients with diabetic retinopathy compared with healthy subjects and patients with T2DM (both P < 0.001, respectively). Also, the periostin level was significantly higher in the PDR group compared to the NPDR group (P = 0.044). Multivariate logistic regression revealed that serum periostin was independently associated with the presence of DR in patients with T2DM (P < 0.001). The receiver operating characteristic (ROC) curves for DR development using serum periostin showed that the area under the receiver operating characteristic curves (AUC) was 0.838 (P < 0.001). CONCLUSIONS The current study demonstrated that serum periostin is significantly associated with the presence of DR in patients with T2DM and is an independent risk factor of DR.
Collapse
Affiliation(s)
- Y Ding
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Q Ge
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - H Qu
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Z Feng
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - J Long
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Q Wei
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Q Zhou
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - R Wu
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - L Yao
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - H Deng
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
39
|
Rizou M, Frangou EA, Marineli F, Prakoura N, Zoidakis J, Gakiopoulou H, Liapis G, Kavvadas P, Chatziantoniou C, Makridakis M, Vlahou A, Boletis J, Vlahakos D, Goumenos D, Daphnis E, Iatrou C, Charonis AS. The family of 14-3-3 proteins and specifically 14-3-3σ are up-regulated during the development of renal pathologies. J Cell Mol Med 2018; 22:4139-4149. [PMID: 29956451 PMCID: PMC6111864 DOI: 10.1111/jcmm.13691] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/19/2018] [Indexed: 12/16/2022] Open
Abstract
Chronic kidney disease, the end result of most renal and some systemic diseases, is a common condition where renal function is compromised due to fibrosis. During renal fibrosis, calreticulin, a multifunctional chaperone of the endoplasmic reticulum (ER) is up‐regulated in tubular epithelial cells (TECs) both in vitro and in vivo. Proteomic analysis of cultured TECs overexpressing calreticulin led to the identification of the family of 14‐3‐3 proteins as key proteins overexpressed as well. Furthermore, an increased expression in the majority of 14‐3‐3 family members was observed in 3 different animal models of renal pathologies: the unilateral ureteric obstruction, the nephrotoxic serum administration and the ischaemia‐reperfusion. In all these models, the 14‐3‐3σ isoform (also known as stratifin) was predominantly overexpressed. As in all these models ischaemia is a common denominator, we showed that the ischaemia‐induced transcription factor HIF1α is specifically associated with the promoter region of the 14‐3‐3σ gene. Finally, we evaluated the expression of the family of 14‐3‐3 proteins and specifically 14‐3‐3σ in biopsies from IgA nephropathy and membranous nephropathy patients. These results propose an involvement of 14‐3‐3σ in renal pathology and provide evidence for the first time that hypoxia may be responsible for its altered expression.
Collapse
Affiliation(s)
- Myrto Rizou
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Eleni A Frangou
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Filio Marineli
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Niki Prakoura
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,Laikon University Hospital, Nephrology Clinic, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Jerome Zoidakis
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Harikleia Gakiopoulou
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, and Laikon Hospital, Athens, Greece
| | - George Liapis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, and Laikon Hospital, Athens, Greece
| | | | | | | | - Antonia Vlahou
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - John Boletis
- Laikon University Hospital, Nephrology Clinic, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Demetrios Vlahakos
- Division of Nephrology, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Dimitrios Goumenos
- Department of Nephrology, Medical School of Patras, University Hospital of Patras, Rio, Greece
| | - Evgenios Daphnis
- Medical School of the University of Crete, University Hospital of Iraklion, Iraklion, Greece
| | - Christos Iatrou
- Center for Nephrology "G. Papadakis", General Hospital of Nikaia-Piraeus, Athens, Greece
| | | |
Collapse
|
40
|
Nordholm A, Mace ML, Gravesen E, Hofman-Bang J, Morevati M, Olgaard K, Lewin E. Klotho and activin A in kidney injury: plasma Klotho is maintained in unilateral obstruction despite no upregulation of Klotho biosynthesis in the contralateral kidney. Am J Physiol Renal Physiol 2018; 314:F753-F762. [PMID: 29187373 PMCID: PMC6031917 DOI: 10.1152/ajprenal.00528.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 11/22/2017] [Accepted: 11/27/2017] [Indexed: 12/17/2022] Open
Abstract
In a new paradigm of etiology related to chronic kidney disease-mineral and bone disorder (CKD-MBD), kidney injury may cause induction of factors in the injured kidney that are released into the circulation and thereby initiate and maintain renal fibrosis and CKD-MBD. Klotho is believed to ameliorate renal fibrosis and CKD-MBD, while activin A might have detrimental effects. The unilateral ureter obstruction (UUO) model is used here to examine this concept by investigating early changes related to renal fibrosis in the obstructed kidney, untouched contralateral kidney, and vasculature which might be affected by secreted factors from the obstructed kidney, and comparing with unilateral nephrectomized controls (UNX). Obstructed kidneys showed early Klotho gene and protein depletion, whereas plasma Klotho increased in both UUO and UNX rats, indicating an altered metabolism of Klotho. Contralateral kidneys had no compensatory upregulation of Klotho and maintained normal expression of the examined fibrosis-related genes, as did remnant UNX kidneys. UUO caused upregulation of transforming growth factor-β and induction of periostin and activin A in obstructed kidneys without changes in the contralateral kidneys. Plasma activin A doubled in UUO rats after 10 days while no changes were seen in UNX rats, suggesting secretion of activin A from the obstructed kidney with potentially systemic effects on CKD-MBD. As such, increased aortic sclerostin was observed in UUO rats compared with UNX and normal controls. The present results are in line with the new paradigm and show very early vascular effects of unilateral kidney fibrosis, supporting the existence of a new kidney-vasculature axis.
Collapse
Affiliation(s)
- Anders Nordholm
- Nephrological Department B, Herlev Hospital, Herlev, University of Copenhagen , Copenhagen , Denmark
| | - Maria L Mace
- Nephrological Department B, Herlev Hospital, Herlev, University of Copenhagen , Copenhagen , Denmark
- Nephrological Department P, Rigshospitalet, Copenhagen , Denmark
| | - Eva Gravesen
- Nephrological Department P, Rigshospitalet, Copenhagen , Denmark
| | | | - Marya Morevati
- Nephrological Department P, Rigshospitalet, Copenhagen , Denmark
| | - Klaus Olgaard
- Nephrological Department P, Rigshospitalet, Copenhagen , Denmark
| | - Ewa Lewin
- Nephrological Department B, Herlev Hospital, Herlev, University of Copenhagen , Copenhagen , Denmark
- Nephrological Department P, Rigshospitalet, Copenhagen , Denmark
| |
Collapse
|
41
|
Actin dynamics at focal adhesions: a common endpoint and putative therapeutic target for proteinuric kidney diseases. Kidney Int 2018; 93:1298-1307. [PMID: 29678354 DOI: 10.1016/j.kint.2017.12.028] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 12/07/2017] [Accepted: 12/13/2017] [Indexed: 01/02/2023]
Abstract
Proteinuria encompasses diverse causes including both genetic diseases and acquired forms such as diabetic and hypertensive nephropathy. The basis of proteinuria is a disturbance in size selectivity of the glomerular filtration barrier, which largely depends on the podocyte: a terminally differentiated epithelial cell type covering the outer surface of the glomerulus. Compromised podocyte structure is one of the earliest signs of glomerular injury. The phenotype of diverse animal models and podocyte cell culture firmly established the essential role of the actin cytoskeleton in maintaining functional podocyte structure. Podocyte foot processes, actin-based membrane extensions, contain 2 molecularly distinct "hubs" that control actin dynamics: a slit diaphragm and focal adhesions. Although loss of foot processes encompasses disassembly of slit diaphragm multiprotein complexes, as long as cells are attached to the glomerular basement membrane, focal adhesions will be the sites in which stress due to filtration flow is counteracted by forces generated by the actin network in foot processes. Numerous studies within last 20 years have identified actin binding and regulatory proteins as well as integrins as essential components of signaling and actin dynamics at focal adhesions in podocytes, suggesting that some of them may become novel, druggable targets for proteinuric kidney diseases. Here we review evidence supporting the idea that current treatments for chronic kidney diseases beneficially and directly target the podocyte actin cytoskeleton associated with focal adhesions and suggest that therapeutic reagents that target the focal adhesion-regulated actin cytoskeleton in foot processes have potential to modernize treatments for chronic kidney diseases.
Collapse
|
42
|
Tang M, Liu B, Bu X, Zhao P. Cross-talk between ovarian cancer cells and macrophages through periostin promotes macrophage recruitment. Cancer Sci 2018. [PMID: 29527764 PMCID: PMC5980394 DOI: 10.1111/cas.13567] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Tumor‐associated macrophages (TAMs) contribute to tumor progression, but it is not clear how they are recruited to tumor sites. Here we showed that periostin (POSTN) was present at high levels in ovarian cancer ascetic fluids and was correlated with CD163+TAMs. The high POSTN level and macrophage infiltration were inversely associated with relapse‐free survival for ovarian cancer patients. In vitro studies showed that coculture with macrophages significantly increased POSTN production in ovarian cancer cells. Further investigation found that POSTN production in ovarian cancer cells was promoted by transforming growth factor‐β generated by macrophages. Moreover, siRNA of POSTN and POSTN neutralizing antibody treatment showed that ovarian cancer cell‐derived POSTN promoted the recruitment of macrophages and modulated their cytokine secretion profile. Collectively, these data indicated that POSTN was an important factor for macrophage recruitment in the tumor microenvironment and is involved in the interactions between macrophages and ovarian cancer cells.
Collapse
Affiliation(s)
- Meng Tang
- Department of Thoracic Surgery, Qingdao Central Hospital, The Second Clinical Hospital of Qingdao University, Qingdao, China
| | - Bingji Liu
- Department of Nuclear Medicine, Qingdao Central Hospital, The Second Clinical Hospital of Qingdao University, Qingdao, China
| | - Xiaocui Bu
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao, China
| | - Peng Zhao
- Biotherapy Center, Qingdao Central Hospital, The Second Clinical Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
43
|
Hatley RJD, Macdonald SJF, Slack RJ, Le J, Ludbrook SB, Lukey PT. An αv-RGD Integrin Inhibitor Toolbox: Drug Discovery Insight, Challenges and Opportunities. Angew Chem Int Ed Engl 2018; 57:3298-3321. [DOI: 10.1002/anie.201707948] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Richard J. D. Hatley
- Fibrosis DPU; Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY UK
| | - Simon J. F. Macdonald
- Fibrosis DPU; Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY UK
| | - Robert J. Slack
- Fibrosis DPU; Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY UK
| | - Joelle Le
- Fibrosis DPU; Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY UK
| | - Steven B. Ludbrook
- Fibrosis DPU; Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY UK
| | - Pauline T. Lukey
- Fibrosis DPU; Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY UK
| |
Collapse
|
44
|
Hatley RJD, Macdonald SJF, Slack RJ, Le J, Ludbrook SB, Lukey PT. Ein Instrumentarium von αv-RGD-Integrin-Inhibitoren: Wirkstoffsuche, Herausforderungen und Möglichkeiten. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201707948] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Richard J. D. Hatley
- Fibrosis and Lung Injury DPU, Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY Großbritannien
| | - Simon J. F. Macdonald
- Fibrosis and Lung Injury DPU, Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY Großbritannien
| | - Robert J. Slack
- Fibrosis and Lung Injury DPU, Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY Großbritannien
| | - Joelle Le
- Fibrosis and Lung Injury DPU, Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY Großbritannien
| | - Steven B. Ludbrook
- Fibrosis and Lung Injury DPU, Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY Großbritannien
| | - Pauline T. Lukey
- Fibrosis and Lung Injury DPU, Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY Großbritannien
| |
Collapse
|
45
|
Renal fibrosis: Recent translational aspects. Matrix Biol 2017; 68-69:318-332. [PMID: 29292218 DOI: 10.1016/j.matbio.2017.12.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/15/2017] [Accepted: 12/21/2017] [Indexed: 01/14/2023]
Abstract
Renal fibrogenesis is the common final pathway to all renal injuries that consequently leads to Chronic Kidney Disease (CKD). Renal fibrogenesis corresponds to the replacement of renal functional tissue by extra-cellular matrix proteins, mainly collagens, that ultimately impairs kidney function. Blockade of the renin angiotensin system by Angiotensin Converting Enzyme inhibitors (ACEi) or Angiotensin Receptor Blockers (ARBs) was the first strategy that proved efficient to blunt the development of renal fibrogenesis independently of its systemic action on blood pressure. Although this strategy has been published 20years ago, there is to date no novel therapeutic targets that are both safe and efficient in hindering renal fibrogenesis and CKD in humans, nor there is any new biomarker to precisely quantify this process. In our review, we will focus on the most recent pathways leading to fibrogenesis which have a high therapeutic potential in humans and on the most promising biomarkers of renal fibrosis.
Collapse
|
46
|
Prakoura N, Chatziantoniou C. Periostin: The Missing Link to Attenuate Kidney Fibrosis. Am J Nephrol 2017. [DOI: 10.1159/000485324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
47
|
Prakoura N, Chatziantoniou C. Periostin in kidney diseases. Cell Mol Life Sci 2017; 74:4315-4320. [PMID: 28884334 PMCID: PMC11107687 DOI: 10.1007/s00018-017-2650-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 09/04/2017] [Indexed: 12/22/2022]
Abstract
Chronic kidney disease is an incurable to date pathology, with renal replacement therapy through dialysis or transplantation being the only available option for end-stage patients. A deeper understanding of the molecular mechanisms governing the progression of kidney diseases will permit the identification of unknown mediators and potential novel markers or targets of therapy which promise more efficient diagnostic and therapeutic applications. Over the last years, periostin was established by several studies as a novel key player in the progression of renal disease. Periostin is de novo expressed focally by the injured kidney cells during the development of renal disease. In diverse cohorts of renal disease patients, the expression levels of periostin in the kidney and urine were highly correlated with the stage of the pathology and the decline of renal function. Subsequent studies in animal models demonstrated that periostin is centrally involved in mediating renal inflammation and fibrosis, contributing to the deterioration of renal structure and function. Genetic or pharmaco-genetic inhibition of periostin in animal models of renal disease was efficient in arresting the progression of the pathology. This review will summarize the recent advances on periostin in the field of kidney diseases and will discuss its utility of as a novel target of therapy for chronic kidney disease.
Collapse
Affiliation(s)
- Niki Prakoura
- Institut National de la Santé Et de la Recherche Médicale UMRS 1155, Tenon Hospital, 4 rue de la Chine, 75020, Paris, France.
| | - Christos Chatziantoniou
- Institut National de la Santé Et de la Recherche Médicale UMRS 1155, Tenon Hospital, 4 rue de la Chine, 75020, Paris, France
- Sorbonne Universités, UPMC Paris 6, Paris, France
| |
Collapse
|
48
|
Prakoura N, Chatziantoniou C. Periostin and Discoidin Domain Receptor 1: New Biomarkers or Targets for Therapy of Renal Disease. Front Med (Lausanne) 2017; 4:52. [PMID: 28536691 PMCID: PMC5422471 DOI: 10.3389/fmed.2017.00052] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 04/20/2017] [Indexed: 12/22/2022] Open
Abstract
Chronic kidney disease (CKD) can be a life-threatening condition, which eventually requires renal replacement therapy through dialysis or transplantation. A lot of effort and resources have been invested the last years in the identification of novel markers of progression and targets for therapy, in order to achieve a more efficient prognosis, diagnosis, and treatment of renal diseases. Using experimental models of renal disease, we identified and studied two promising candidates: periostin, a matricellular protein with high expression in bone and dental tissues, and discoidin domain receptor 1 (DDR1), a transmembrane collagen receptor of the tyrosine kinase family. Both proteins are inactive in physiological conditions, while they are highly upregulated during development of renal disease and are primarily expressed at the sites of injury. Further studies demonstrated that both periostin and DDR1 are involved in the regulation of inflammation and fibrosis, two major processes implicated in the development of renal disease. Targeting of either protein by genetic deletion or pharmacogenetic inhibition via antisense oligonucleotides highly attenuates renal damage and preserves renal structure and function in several animal models. The scope of this review is to summarize the existing evidence supporting the role of periostin and DDR1 as novel biomarkers and therapeutic targets in CKD.
Collapse
Affiliation(s)
- Niki Prakoura
- Institut National de la Santé Et de la Recherche Médicale UMRS 1155, Tenon Hospital, Paris, France
| | - Christos Chatziantoniou
- Institut National de la Santé Et de la Recherche Médicale UMRS 1155, Tenon Hospital, Paris, France.,Sorbonne Universités, UPMC Paris 6, Paris, France
| |
Collapse
|
49
|
Prakoura N, Chatziantoniou C. Matricellular Proteins and Organ Fibrosis. CURRENT PATHOBIOLOGY REPORTS 2017. [DOI: 10.1007/s40139-017-0138-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|