1
|
Dai Y, Li Y, Xu J, Zhang J. A highly selective inhibitor of discoidin domain receptor-1 (DDR1-IN-1) protects corneal epithelial cells from YAP/ACSL4-mediated ferroptosis in dry eye. Br J Pharmacol 2024; 181:4245-4261. [PMID: 38978400 DOI: 10.1111/bph.16491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/03/2024] [Accepted: 05/26/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND AND PURPOSE This study investigated the involvement of discoidin domain receptor (DDR) in dry eye and assessed the potential of specific DDR inhibitors as a therapeutic strategy for dry eye by exploring the underlying mechanism. EXPERIMENTAL APPROACH Dry eye was induced in Wistar rats by applying 0.2% benzalkonium chloride (BAC), after which rats were treated topically for 7 days with DDR1-IN-1, a selective inhibitor of DDR1. Clinical manifestations of dry eye were assessed on Day-7 post-treatment. Histological evaluation of corneal damage was performed using haematoxylin and eosin (H&E) staining. In vitro, immortalized human corneal epithelial cells (HCECs) exposed to hyperosmotic stress (HS) were treated with varying doses of DDR1-IN-1 for 24 h. The levels of lipid peroxidation in dry eye corneas or HS-stimulated HCECs were assessed. Protein levels of DDR1/DDR2 and related pathways were detected by western blotting. The cellular distribution of acyl-CoA synthetase long chain family member 4 (ACSL4) and Yes-associated protein (YAP) was evaluated using immunohistochemistry or immunofluorescent staining. KEY RESULTS In dry eye corneas, only DDR1 expression was significantly up-regulated compared with normal controls. DDR1-IN-1 treatment significantly alleviated dry eye symptoms in vivo. The treatment remarkably reduced lipid hydroperoxide (LPO) levels and suppressed the expression of ferroptosis markers, particularly ACSL4. Overexpression or reactivation of YAP diminished the protective effects of DDR1-IN-1, indicating the involvement of the Hippo/YAP pathway in DDR1-targeted therapeutic effects. CONCLUSIONS AND IMPLICATIONS This study confirms the significance of DDR1 in dry eye and highlights the potential of selective DDR1 inhibitor(s) for dry eye treatment.
Collapse
Affiliation(s)
- Yiqin Dai
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yue Li
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Jianjiang Xu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Jing Zhang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| |
Collapse
|
2
|
Zhang M, Barroso E, Peña L, Rada P, Valverde ÁM, Wahli W, Palomer X, Vázquez-Carrera M. PPARβ/δ attenuates hepatic fibrosis by reducing SMAD3 phosphorylation and p300 levels via AMPK in hepatic stellate cells. Biomed Pharmacother 2024; 179:117303. [PMID: 39153437 DOI: 10.1016/j.biopha.2024.117303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/04/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024] Open
Abstract
The role of peroxisome proliferator-activated receptor (PPAR)β/δ in hepatic fibrosis remains a subject of debate. Here, we examined the effects of a PPARβ/δ agonist on the pathogenesis of liver fibrosis and the activation of hepatic stellate cells (HSCs), the main effector cells in liver fibrosis, in response to the pro-fibrotic stimulus transforming growth factor-β (TGF-β). The PPARβ/δ agonist GW501516 completely prevented glucose intolerance and peripheral insulin resistance, blocked the accumulation of collagen in the liver, and attenuated the expression of inflammatory and fibrogenic genes in mice fed a choline-deficient high-fat diet (CD-HFD). The antifibrogenic effect of GW501516 observed in the livers CD-HFD-fed mice could occur through an action on HSCs since primary HSCs isolated from Ppard-/- mice showed increased mRNA levels of the profibrotic gene Col1a1. Moreover, PPARβ/δ activation abrogated TGF-β1-mediated cell migration (an indicator of cell activation) in LX-2 cells (immortalized activated human HSCs). Likewise, GW501516 attenuated the phosphorylation of the main downstream intracellular protein target of TGF-β1, suppressor of mothers against decapentaplegic (SMAD)3, as well as the levels of the SMAD3 co-activator p300 via the activation of AMP-activated protein kinase (AMPK) and the subsequent inhibition of extracellular signal-regulated kinase-1/2 (ERK1/2) in LX-2 cells. Overall, these findings uncover a new mechanism by which the activation of AMPK by a PPARβ/δ agonist reduces TGF-β1-mediated activation of HSCs and fibrosis via the reduction of both SMAD3 phosphorylation and p300 levels.
Collapse
Affiliation(s)
- Meijian Zhang
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona 08028, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid 28029, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat 08950, Spain
| | - Emma Barroso
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona 08028, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid 28029, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat 08950, Spain.
| | - Lucía Peña
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona 08028, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid 28029, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat 08950, Spain
| | - Patricia Rada
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid 28029, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC/UAM), Madrid, Spain
| | - Ángela M Valverde
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid 28029, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC/UAM), Madrid, Spain
| | - Walter Wahli
- Center for Integrative Genomics, University of Lausanne, Lausanne CH-1015, Switzerland; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; ToxAlim (Research Center in Food Toxicology), INRAE, UMR1331, Toulouse Cedex F-31300, France
| | - Xavier Palomer
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona 08028, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid 28029, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat 08950, Spain
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona 08028, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid 28029, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat 08950, Spain.
| |
Collapse
|
3
|
Abbad L, Détrait M, Kavvadas P, Bergonnier D, Melis L, Laudette M, Migeon T, Verpont MC, Lucas A, Chatziantoniou C, Lezoualc'h F. Signaling through cAMP-Epac1 induces metabolic reprogramming to protect podocytes in glomerulonephritis. Kidney Int 2024; 106:450-469. [PMID: 38821447 DOI: 10.1016/j.kint.2024.05.010] [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: 09/22/2023] [Revised: 03/27/2024] [Accepted: 05/17/2024] [Indexed: 06/02/2024]
Abstract
Unlike classical protein kinase A, with separate catalytic and regulatory subunits, EPACs are single chain multi-domain proteins containing both catalytic and regulatory elements. The importance of cAMP-Epac-signaling as an energy provider has emerged over the last years. However, little is known about Epac1 signaling in chronic kidney disease. Here, we examined the role of Epac1 during the progression of glomerulonephritis (GN). We first observed that total genetic deletion of Epac1 in mice accelerated the progression of nephrotoxic serum (NTS)-induced GN. Next, mice with podocyte-specific conditional deletion of Epac1 were generated and showed that NTS-induced GN was exacerbated in these mice. Gene expression analysis in glomeruli at the early and late phases of GN showed that deletion of Epac1 in podocytes was associated with major alterations in mitochondrial and metabolic processes and significant dysregulation of the glycolysis pathway. In vitro, Epac1 activation in a human podocyte cell line increased mitochondrial function to cope with the extra energy demand under conditions of stress. Furthermore, Epac1-induced glycolysis and lactate production improved podocyte viability. To verify the in vivo therapeutic potential of Epac1 activation, the Epac1 selective cAMP mimetic 8-pCPT was administered in wild type mice after induction of GN. 8-pCPT alleviated the progression of GN by improving kidney function with decreased structural injury with decreased crescent formation and kidney inflammation. Importantly, 8-pCPT had no beneficial effect in mice with Epac1 deletion in podocytes. Thus, our data suggest that Epac1 activation is an essential protective mechanism in GN by reprogramming podocyte metabolism. Hence, targeting Epac1 activation could represent a potential therapeutic approach.
Collapse
Affiliation(s)
- Lilia Abbad
- INSERM UMR S 1155, Common and Rare Kidney Diseases, Tenon Hospital, Faculty of Health, Sorbonne University, Paris, France
| | - Maximin Détrait
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM, University Toulouse III-Paul Sabatier, UMR 1297-I2MC, Toulouse, France
| | - Panagiotis Kavvadas
- INSERM UMR S 1155, Common and Rare Kidney Diseases, Tenon Hospital, Faculty of Health, Sorbonne University, Paris, France
| | - Dorian Bergonnier
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM, University Toulouse III-Paul Sabatier, UMR 1297-I2MC, Toulouse, France
| | - Lisa Melis
- INSERM UMR S 1155, Common and Rare Kidney Diseases, Tenon Hospital, Faculty of Health, Sorbonne University, Paris, France
| | - Marion Laudette
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM, University Toulouse III-Paul Sabatier, UMR 1297-I2MC, Toulouse, France
| | - Tiffany Migeon
- INSERM UMR S 1155, Common and Rare Kidney Diseases, Tenon Hospital, Faculty of Health, Sorbonne University, Paris, France
| | - Marie-Christine Verpont
- INSERM UMR S 1155, Common and Rare Kidney Diseases, Tenon Hospital, Faculty of Health, Sorbonne University, Paris, France
| | - Alexandre Lucas
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM, University Toulouse III-Paul Sabatier, UMR 1297-I2MC, Toulouse, France
| | - Christos Chatziantoniou
- INSERM UMR S 1155, Common and Rare Kidney Diseases, Tenon Hospital, Faculty of Health, Sorbonne University, Paris, France.
| | - Frank Lezoualc'h
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM, University Toulouse III-Paul Sabatier, UMR 1297-I2MC, Toulouse, France.
| |
Collapse
|
4
|
Mariadoss AVA, Wang CZ. Exploring the Cellular and Molecular Mechanism of Discoidin Domain Receptors (DDR1 and DDR2) in Bone Formation, Regeneration, and Its Associated Disease Conditions. Int J Mol Sci 2023; 24:14895. [PMID: 37834343 PMCID: PMC10573612 DOI: 10.3390/ijms241914895] [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: 09/06/2023] [Revised: 10/01/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023] Open
Abstract
The tyrosine kinase family receptor of discoidin domain receptors (DDR1 and DDR2) is known to be activated by extracellular matrix collagen catalytic binding protein receptors. They play a remarkable role in cell proliferation, differentiation, migration, and cell survival. DDR1 of the DDR family regulates matrix-metalloproteinase, which causes extracellular matrix (ECM) remodeling and reconstruction during unbalanced homeostasis. Collagenous-rich DDR1 triggers the ECM of cartilage to regenerate the cartilage tissue in osteoarthritis (OA) and temporomandibular disorder (TMD). Moreover, DDR2 is prominently present in the fibroblasts, smooth muscle cells, myofibroblasts, and chondrocytes. It is crucial in generating and breaking collagen vital cellular activities like proliferation, differentiation, and adhesion mechanisms. However, the deficiency of DDR1 rather than DDR2 was detrimental in cases of OA and TMDs. DDR1 stimulated the ECM cartilage and improved bone regeneration. Based on the above information, we made an effort to outline the advancement of the utmost promising DDR1 and DDR2 regulation in bone and cartilage, also summarizing their structural, biological activity, and selectivity.
Collapse
Affiliation(s)
| | - Chau-Zen Wang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- College of Professional Studies, National Pingtung University of Science and Technology, Pingtung 912301, Taiwan
| |
Collapse
|
5
|
Li X, Chen H, Zhang D. Discoidin domain receptor 1 may be involved in biological barrier homeostasis. J Clin Pharm Ther 2022; 47:2397-2407. [PMID: 35665520 DOI: 10.1111/jcpt.13705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 04/08/2022] [Accepted: 04/25/2022] [Indexed: 12/24/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE Discoidin domain receptor 1 (DDR1) is a receptor tyrosine kinase involved in the pathological processes of several diseases, such as keloid formation, renal fibrosis, atherosclerosis, tumours, and inflammatory processes. The biological barrier is the first line of defence against pathogens, and its disruption is closely related to diseases. In this review, we attempt to elucidate the relationship between DDR1 and the biological barrier, explore the potential biological value of DDR1, and review the current research status and clinical potential of DDR1-selective inhibitors. METHODS We conducted an extensive literature search on PubMed to collect studies on the relevance of DDR1 to biological barriers and DDR1-selective inhibitors. With these studies, we explored the relationship between DDR1 and biological barriers and briefly reviewed representative DDR1-selective inhibitors that have been reported in recent years. RESULTS AND DISCUSSION First, the review of the potential mechanisms by which DDR1 regulates biological barriers, including the epithelial, vascular, glomerular filtration, blood-labyrinth, and blood-brain barriers. In the body, DDR1 dysfunction and aberrant expression may be involved in the homeostasis of the biological barrier. Secondly, the review of DDR1 inhibitors reported in recent years shows that DDR1-targeted inhibition is an attractive and promising pharmacological intervention. WHAT IS NEW AND CONCLUSIONS This review shows that DDR1 is involved in various physiological and pathological processes and in the regulation of biological barrier homeostasis. However, studies on DDR1 and biological barriers are still scarce, and further studies are needed to elucidate their specific mechanisms. The development of targeted inhibitors provides a new direction and idea to study the mechanism of DDR1.
Collapse
Affiliation(s)
- Xiaoli Li
- Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Huiling Chen
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Dekui Zhang
- Department of Gastroenterology, Key Laboratory of Digestive Diseases, LanZhou University Second Hospital, LanZhou University, Lanzhou, China
| |
Collapse
|
6
|
Zhao W, He C, Jiang J, Zhao Z, Yuan H, Wang F, Shen B. The role of discoid domain receptor 1 on renal tubular epithelial pyroptosis in diabetic nephropathy. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2022; 26:427-438. [PMID: 36302618 PMCID: PMC9614395 DOI: 10.4196/kjpp.2022.26.6.427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 04/13/2022] [Accepted: 04/13/2022] [Indexed: 11/07/2022]
Abstract
Pyroptosis, a form of cell death associated with inflammation, is known to be involved in diabetic nephropathy (DN), and discoid domain receptor 1 (DDR1), an inflammatory regulatory protein, is reported to be associated with diabetes. However, the mechanism underlying DDR1 regulation and pyroptosis in DN remains unknown. We aimed to investigate the effect of DDR1 on renal tubular epithelial cell pyroptosis and the mechanism underlying DN. In this study, we used high glucose (HG)-treated HK-2 cells and rats with a single intraperitoneal injection of streptozotocin as DN models. Subsequently, the expression of pyroptosis-related proteins (cleaved caspase-1, GSDMD-N, Interleukin-1β [IL-1β], and interleukin-18 [IL-18]), DDR1, phosphorylated NF-κB (p-NF-κB), and NLR family pyrin domain-containing 3 (NLRP3) inflammasomes were determined through Western blotting. IL-1β and IL-18 levels were determined using ELISA. The rate of pyroptosis was assessed by propidium iodide (PI) staining. The results revealed upregulated expression of pyroptosis-related proteins and increased concentration of IL-1β and IL-18, accompanied by DDR1, p-NF-κB, and NLRP3 upregulation in DN rat kidney tissues and HG-treated HK-2 cells. Moreover, DDR1 knockdown in the background of HG treatment resulted in inhibited expression of pyroptosis-related proteins and attenuation of IL-1β and IL-18 production and PI-positive cell frequency via the NF-κB/NLRP3 pathway in HK-2 cells. However, NLRP3 overexpression reversed the effect of DDR1 knockdown on pyroptosis. In conclusion, we demonstrated that DDR1 may be associated with pyroptosis, and DDR1 knockdown inhibited HG-induced renal tubular epithelial cell pyroptosis. The NF-κB/NLRP3 pathway is probably involved in the underlying mechanism of these findings.
Collapse
Affiliation(s)
- Weichen Zhao
- Department of Pharmacy, Lu'an Hospital Affiliated to Anhui Medical University, Lu'an People's Hospital, Lu'an, Anhui 237005, China
| | - Chunyuan He
- Department of Pharmacy, Lu'an Hospital Affiliated to Anhui Medical University, Lu'an People's Hospital, Lu'an, Anhui 237005, China
| | - Junjie Jiang
- Department of Pharmacy, Lu'an Hospital Affiliated to Anhui Medical University, Lu'an People's Hospital, Lu'an, Anhui 237005, China
| | - Zongbiao Zhao
- Department of Pharmacy, Lu'an Hospital Affiliated to Anhui Medical University, Lu'an People's Hospital, Lu'an, Anhui 237005, China
| | - Hongzhong Yuan
- Department of Pharmacy, Lu'an Hospital Affiliated to Anhui Medical University, Lu'an People's Hospital, Lu'an, Anhui 237005, China
| | - Facai Wang
- Department of Pharmacy, Lu'an Hospital Affiliated to Anhui Medical University, Lu'an People's Hospital, Lu'an, Anhui 237005, China
| | - Bingxiang Shen
- Department of Pharmacy, Lu'an Hospital Affiliated to Anhui Medical University, Lu'an People's Hospital, Lu'an, Anhui 237005, China
| |
Collapse
|
7
|
Borza CM, Bolas G, Pozzi A. Genetic and pharmacological tools to study the role of discoidin domain receptors in kidney disease. Front Pharmacol 2022; 13:1001122. [PMID: 36249782 PMCID: PMC9554349 DOI: 10.3389/fphar.2022.1001122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Following injury the kidney undergoes a repair process, which results in replacement of the injured tissue with little evidence of damage. However, repetitive injuries or inability of the kidney to stop the repair process result in abnormal deposition of extracellular matrix (ECM) components leading to fibrosis and organ dysfunction. The synthesis/degradation of ECM components is finely regulated by several factors, including discoidin domain receptors (DDRs). These are receptor tyrosine kinases that are activated by collagens. Upon activation, DDRs control several cell functions that, when exacerbated, contribute to kidney injury and fibrosis. DDRs are undetectable in healthy kidney, but become rapidly upregulated in several kidney fibrotic conditions, thus making them attractive anti-fibrotic targets. DDRs contribute to kidney injury and fibrosis by promoting apoptosis of injured kidney cells, stimulating the production of pro-inflammatory cytokines, and regulating the production of ECM components. They achieve these effects by activating canonical intracellular molecules or by directly interacting with nuclear chromatin and promoting the transcription of pro-fibrotic genes. The goal of this review is to highlight canonical and non-canonical mechanisms whereby DDRs contribute to kidney injury/fibrosis. This review will summarize key findings obtained using cells and mice lacking DDRs and it will discuss the discovery and development of targeted DDR small molecule- and antisense-based inhibitors. Understanding the molecular mechanisms whereby DDRs control kidney injury and fibrosis might enable us to not only develop more selective and potent inhibitors, but to also determine when DDR inhibition needs to be achieved to prevent and/or halt the development of kidney fibrosis.
Collapse
Affiliation(s)
- Corina M. Borza
- Department of Medicine (Division of Nephrology), Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Gema Bolas
- Department of Medicine (Division of Nephrology), Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Ambra Pozzi
- Department of Medicine (Division of Nephrology), Vanderbilt University School of Medicine, Nashville, TN, United States
- Veterans Affairs Hospitals, Nashville, TN, United States
| |
Collapse
|
8
|
Madison J, Wilhelm K, Meehan DT, Delimont D, Samuelson G, Cosgrove D. Glomerular basement membrane deposition of collagen α1(III) in Alport glomeruli by mesangial filopodia injures podocytes via aberrant signaling through DDR1 and integrin α2β1. J Pathol 2022; 258:26-37. [PMID: 35607980 PMCID: PMC9378723 DOI: 10.1002/path.5969] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/29/2022] [Accepted: 05/20/2022] [Indexed: 11/20/2022]
Abstract
In Alport mice, activation of the endothelin A receptor (ETA R) in mesangial cells results in sub-endothelial invasion of glomerular capillaries by mesangial filopodia. Filopodia deposit mesangial matrix in the glomerular basement membrane (GBM), including laminin 211 which activates NF-κB, resulting in induction of inflammatory cytokines. Herein we show that collagen α1(III) is also deposited in the GBM. Collagen α1(III) localized to the mesangium in wild-type mice and was found in both the mesangium and the GBM in Alport mice. We show that collagen α1(III) activates discoidin domain receptor family, member 1 (DDR1) receptors both in vitro and in vivo. To elucidate whether collagen α1(III) might cause podocyte injury, cultured murine Alport podocytes were overlaid with recombinant collagen α1(III), or not, for 24 h and RNA was analyzed by RNA sequencing (RNA-seq). These same cells were subjected to siRNA knockdown for integrin α2 or DDR1 and the RNA was analyzed by RNA-seq. Results were validated in vivo using RNA-seq from RNA isolated from wild-type and Alport mouse glomeruli. Numerous genes associated with podocyte injury were up- or down-regulated in both Alport glomeruli and cultured podocytes treated with collagen α1(III), 18 of which have been associated previously with podocyte injury or glomerulonephritis. The data indicate α2β1 integrin/DDR1 co-receptor signaling as the dominant regulatory mechanism. This may explain earlier studies where deletion of either DDR1 or α2β1 integrin in Alport mice ameliorates renal pathology. © 2022 Boys Town National Research Hospital. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
|
9
|
Borza CM, Bolas G, Bock F, Zhang X, Akabogu FC, Zhang MZ, de Caestecker M, Yang M, Yang H, Lee E, Gewin L, Fogo AB, McDonald WH, Zent R, Pozzi A. DDR1 contributes to kidney inflammation and fibrosis by promoting the phosphorylation of BCR and STAT3. JCI Insight 2021; 7:150887. [PMID: 34941574 PMCID: PMC8855801 DOI: 10.1172/jci.insight.150887] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 12/21/2021] [Indexed: 11/25/2022] Open
Abstract
Discoidin domain receptor 1 (DDR1), a receptor tyrosine kinase activated by collagen, contributes to chronic kidney disease. However, its role in acute kidney injury and subsequent development of kidney fibrosis is not clear. Thus, we performed a model of severe ischemia/reperfusion-induced acute kidney injury that progressed to kidney fibrosis in WT and Ddr1-null mice. We showed that Ddr1-null mice had reduced acute tubular injury, inflammation, and tubulointerstitial fibrosis with overall decreased renal monocyte chemoattractant protein (MCP-1) levels and STAT3 activation. We identified breakpoint cluster region (BCR) protein as a phosphorylated target of DDR1 that controls MCP-1 production in renal proximal tubule epithelial cells. DDR1-induced BCR phosphorylation or BCR downregulation increased MCP-1 secretion, suggesting that BCR negatively regulates the levels of MCP-1. Mechanistically, phosphorylation or downregulation of BCR increased β-catenin activity and in turn MCP-1 production. Finally, we showed that DDR1-mediated STAT3 activation was required to stimulate the secretion of TGF-β. Thus, DDR1 contributes to acute and chronic kidney injury by regulating BCR and STAT3 phosphorylation and in turn the production of MCP-1 and TGF-β. These findings identify DDR1 an attractive therapeutic target for ameliorating both proinflammatory and profibrotic signaling in kidney disease.
Collapse
Affiliation(s)
- Corina M. Borza
- Department of Medicine, Division of Nephrology and Hypertension, and
| | - Gema Bolas
- Department of Medicine, Division of Nephrology and Hypertension, and
| | - Fabian Bock
- Department of Medicine, Division of Nephrology and Hypertension, and
| | - Xiuqi Zhang
- Department of Medicine, Division of Nephrology and Hypertension, and
| | - Favour C. Akabogu
- Department of Medicine, Division of Nephrology and Hypertension, and
| | - Ming-Zhi Zhang
- Department of Medicine, Division of Nephrology and Hypertension, and
| | | | - Min Yang
- Department of Medicine, Division of Nephrology and Hypertension, and
| | - Haichun Yang
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Ethan Lee
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Leslie Gewin
- Division of Nephrology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Agnes B. Fogo
- Department of Medicine, Division of Nephrology and Hypertension, and
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - W. Hayes McDonald
- Proteomics Laboratory, Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee, USA
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Roy Zent
- Department of Medicine, Division of Nephrology and Hypertension, and
- Department of Veterans Affairs, Nashville, Nashville, Tennessee, USA
| | - Ambra Pozzi
- Department of Medicine, Division of Nephrology and Hypertension, and
- Department of Veterans Affairs, Nashville, Nashville, Tennessee, USA
| |
Collapse
|
10
|
Sannomiya Y, Kaseda S, Kamura M, Yamamoto H, Yamada H, Inamoto M, Kuwazuru J, Niino S, Shuto T, Suico MA, Kai H. The role of discoidin domain receptor 2 in the renal dysfunction of alport syndrome mouse model. Ren Fail 2021; 43:510-519. [PMID: 33706638 PMCID: PMC7971217 DOI: 10.1080/0886022x.2021.1896548] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 02/09/2021] [Accepted: 02/19/2021] [Indexed: 02/08/2023] Open
Abstract
Alport syndrome (AS) is a hereditary glomerular nephritis caused by mutation in one of the type IV collagen genes α3/α4/α5 that encode the heterotrimer COL4A3/4/5. Failure to form a heterotrimer due to mutation leads to the dysfunction of the glomerular basement membrane, and end-stage renal disease. Previous reports have suggested the involvement of the receptor tyrosine kinase discoidin domain receptor (DDR) 1 in the progression of AS pathology. However, due to the similarity between DDR1 and DDR2, the role of DDR2 in AS pathology is unclear. Here, we investigated the involvement of DDR2 in AS using the X-linked AS mouse model. Mice were treated subcutaneously with saline or antisense oligonucleotide (ASO; 5 mg/kg or 15 mg/kg per week) for 8 weeks. Renal function parameters and renal histology were analyzed, and the gene expressions of inflammatory cytokines were determined in renal tissues. The expression level of DDR2 was highly elevated in kidney tissues of AS mice. Knockdown of Ddr2 using Ddr2-specific ASO decreased the Ddr2 expression. However, the DDR2 ASO treatment did not improve the proteinuria or decrease the BUN level. DDR2 ASO also did not significantly ameliorate the renal injury, inflammation and fibrosis in AS mice. These results showed that Ddr2 knockdown by ASO had no notable effect on the progression of AS indicating that DDR2 may not be critically involved in AS pathology. This finding may provide useful information and further understanding of the role of DDRs in AS.
Collapse
Affiliation(s)
- Yuya Sannomiya
- Department of Molecular Medicine Graduate School of Pharmaceutical Sciences, Kumamoto, Japan
| | - Shota Kaseda
- Department of Molecular Medicine Graduate School of Pharmaceutical Sciences, Kumamoto, Japan
- Program for Leading Graduate Schools “HIGO (Health life science: Interdisciplinary and Glocal Oriented) Program”, Kumamoto University, Kumamoto, Japan
| | - Misato Kamura
- Department of Molecular Medicine Graduate School of Pharmaceutical Sciences, Kumamoto, Japan
- Program for Leading Graduate Schools “HIGO (Health life science: Interdisciplinary and Glocal Oriented) Program”, Kumamoto University, Kumamoto, Japan
| | | | | | | | - Jun Kuwazuru
- Department of Molecular Medicine Graduate School of Pharmaceutical Sciences, Kumamoto, Japan
| | - Saki Niino
- Department of Molecular Medicine Graduate School of Pharmaceutical Sciences, Kumamoto, Japan
| | - Tsuyoshi Shuto
- Department of Molecular Medicine Graduate School of Pharmaceutical Sciences, Kumamoto, Japan
- Global Center for Natural Resources Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Mary Ann Suico
- Department of Molecular Medicine Graduate School of Pharmaceutical Sciences, Kumamoto, Japan
- Global Center for Natural Resources Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hirofumi Kai
- Department of Molecular Medicine Graduate School of Pharmaceutical Sciences, Kumamoto, Japan
- Program for Leading Graduate Schools “HIGO (Health life science: Interdisciplinary and Glocal Oriented) Program”, Kumamoto University, Kumamoto, Japan
- Global Center for Natural Resources Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
11
|
Denny WA, Flanagan JU. Inhibitors of Discoidin Domain Receptor (DDR) Kinases for Cancer and Inflammation. Biomolecules 2021; 11:1671. [PMID: 34827669 PMCID: PMC8615839 DOI: 10.3390/biom11111671] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 01/22/2023] Open
Abstract
The discoidin domain receptor tyrosine kinases DDR1 and DDR2 are distinguished from other kinase enzymes by their extracellular domains, which interact with collagen rather than with peptidic growth factors, before initiating signaling via tyrosine phosphorylation. They share significant sequence and structural homology with both the c-Kit and Bcr-Abl kinases, and so many inhibitors of those kinases are also effective. Nevertheless, there has been an extensive research effort to develop potent and specific DDR inhibitors. A key interaction for many of these compounds is H-bonding to Met-704 in a hydrophobic pocket of the DDR enzyme. The most widespread use of DDR inhibitors has been for cancer therapy, but they have also shown effectiveness in animal models of inflammatory conditions such as Alzheimer's and Parkinson's diseases, and in chronic renal failure and glomerulonephritis.
Collapse
Affiliation(s)
- William A. Denny
- Auckland Cancer Society Research Centre, Maurice Wilkins Centre, School of Medical Sciences, University of Auckland, Auckland 1142, New Zealand;
| | - Jack U. Flanagan
- Auckland Cancer Society Research Centre, Maurice Wilkins Centre, School of Medical Sciences, University of Auckland, Auckland 1142, New Zealand;
- Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, University of Auckland, Auckland 1142, New Zealand
| |
Collapse
|
12
|
Hayuningtyas RA, Han M, Choi S, Kwak MS, Park IH, Lee JH, Choi JE, Kim DK, Son M, Shin JS. The collagen structure of C1q induces wound healing by engaging discoidin domain receptor 2. Mol Med 2021; 27:125. [PMID: 34602056 PMCID: PMC8489103 DOI: 10.1186/s10020-021-00388-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 09/24/2021] [Indexed: 01/01/2023] Open
Abstract
Background C1q has been reported to reveal complement-independent roles in immune and non-immune cells. C1q binds to its specific receptors to regulate distinct functions that rely on the environment and cell types. Discoidin domain receptor 2 (DDR2) is activated by collagen and functions in wound healing by controlling matrix metalloproteinase (MMP) expression. Since C1q exhibits a collagen-like structure, we hypothesized that C1q might engage DDR2 to regulate wound healing and extracellular matrix (ECM) remodeling. Methods Cell-based assay, proximity ligation assay, ELISA, and surface plasmon analysis were utilized to investigate DDR2 and C1q binding. We also investigate the C1q-mediated in vitro wound healing ability using the human fibrosarcoma cell line, HT1080. Results C1q induced the phosphorylation of DDR2, p38 kinase, and ERK1/2. C1q and DDR2 binding improved cell migration and induced MMP2 and MMP9 expression. DDR2-specific shRNA reduced C1q-mediated cell migration for wound healing. Conclusions C1q is a new DDR2 ligand that promotes wound healing. These findings have therapeutic implications in wound healing-related diseases.
Collapse
Affiliation(s)
- Ria Aryani Hayuningtyas
- Department of Microbiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, Republic of Korea.,Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Myeonggil Han
- Department of Microbiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, Republic of Korea.,Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Seoyeon Choi
- Department of Microbiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, Republic of Korea.,Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Man Sup Kwak
- Department of Microbiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - In Ho Park
- Severance Biomedical Science Institute and Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Ji-Hyun Lee
- Department of Immunology and Institute for Medical Sciences, Jeonbuk National University, Medical School, Jeonju, Jeollabuk-do, 54907, Republic of Korea
| | - Ji Eun Choi
- Department of Pediatrics, Seoul National University Boramae Hospital, Seoul National University College of Medicine, Seoul, 07061, Republic of Korea
| | - Dae Ki Kim
- Department of Immunology and Institute for Medical Sciences, Jeonbuk National University, Medical School, Jeonju, Jeollabuk-do, 54907, Republic of Korea
| | - Myoungsun Son
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA. .,Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11549, USA.
| | - Jeon-Soo Shin
- Department of Microbiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, Republic of Korea. .,Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea. .,Severance Biomedical Science Institute and Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| |
Collapse
|
13
|
Interplay between extracellular matrix components and cellular and molecular mechanisms in kidney fibrosis. Clin Sci (Lond) 2021; 135:1999-2029. [PMID: 34427291 DOI: 10.1042/cs20201016] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022]
Abstract
Chronic kidney disease (CKD) is characterized by pathological accumulation of extracellular matrix (ECM) proteins in renal structures. Tubulointerstitial fibrosis is observed in glomerular diseases as well as in the regeneration failure of acute kidney injury (AKI). Therefore, finding antifibrotic therapies comprises an intensive research field in Nephrology. Nowadays, ECM is not only considered as a cellular scaffold, but also exerts important cellular functions. In this review, we describe the cellular and molecular mechanisms involved in kidney fibrosis, paying particular attention to ECM components, profibrotic factors and cell-matrix interactions. In response to kidney damage, activation of glomerular and/or tubular cells may induce aberrant phenotypes characterized by overproduction of proinflammatory and profibrotic factors, and thus contribute to CKD progression. Among ECM components, matricellular proteins can regulate cell-ECM interactions, as well as cellular phenotype changes. Regarding kidney fibrosis, one of the most studied matricellular proteins is cellular communication network-2 (CCN2), also called connective tissue growth factor (CTGF), currently considered as a fibrotic marker and a potential therapeutic target. Integrins connect the ECM proteins to the actin cytoskeleton and several downstream signaling pathways that enable cells to respond to external stimuli in a coordinated manner and maintain optimal tissue stiffness. In kidney fibrosis, there is an increase in ECM deposition, lower ECM degradation and ECM proteins cross-linking, leading to an alteration in the tissue mechanical properties and their responses to injurious stimuli. A better understanding of these complex cellular and molecular events could help us to improve the antifibrotic therapies for CKD.
Collapse
|
14
|
Inhibition of discoidin domain receptors by imatinib prevented pancreatic fibrosis demonstrated in experimental chronic pancreatitis model. Sci Rep 2021; 11:12894. [PMID: 34145346 PMCID: PMC8213731 DOI: 10.1038/s41598-021-92461-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 06/10/2021] [Indexed: 12/23/2022] Open
Abstract
Discoidin domain receptors (DDR1 and DDR2) are the collagen receptors of the family tyrosine kinases, which play significant role in the diseases like inflammation, fibrosis and cancer. Chronic pancreatitis (CP) is a fibro-inflammatory disease in which recurrent pancreatic inflammation leads to pancreatic fibrosis. In the present study, we have investigated the role of DDR1 and DDR2 in CP. The induced expression of DDR1 and DDR2 was observed in primary pancreatic stellate cells (PSCs) and cerulein-induced CP. Subsequently, the protective effects of DDR1/DDR2 inhibitor, imatinib (IMT) were investigated. Pharmacological intervention with IMT effectively downregulated DDR1 and DDR2 expression. Further, IMT treatment reduced pancreatic injury, inflammation, extracellular matrix deposition and PSCs activation along with inhibition of TGF-β1/Smad signaling pathway. Taken together, these results suggest that inhibition of DDR1 and DDR2 controls pancreatic inflammation and fibrosis, which could represent an attractive and promising therapeutic strategy for the treatment of CP.
Collapse
|
15
|
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
|
16
|
Kim JJ, David JM, Wilbon SS, Santos JV, Patel DM, Ahmad A, Mitrofanova A, Liu X, Mallela SK, Ducasa GM, Ge M, Sloan AJ, Al-Ali H, Boulina M, Mendez AJ, Contreras GN, Prunotto M, Sohail A, Fridman R, Miner JH, Merscher S, Fornoni A. Discoidin domain receptor 1 activation links extracellular matrix to podocyte lipotoxicity in Alport syndrome. EBioMedicine 2020; 63:103162. [PMID: 33340991 PMCID: PMC7750578 DOI: 10.1016/j.ebiom.2020.103162] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/21/2020] [Accepted: 11/24/2020] [Indexed: 12/11/2022] Open
Abstract
Background Discoidin domain receptor 1 (DDR1) is a receptor tyrosine kinase that is activated by collagens that is involved in the pathogenesis of fibrotic disorders. Interestingly, de novo production of the collagen type I (Col I) has been observed in Col4a3 knockout mice, a mouse model of Alport Syndrome (AS mice). Deletion of the DDR1 in AS mice was shown to improve survival and renal function. However, the mechanisms driving DDR1-dependent fibrosis remain largely unknown. Methods Podocyte pDDR1 levels, Collagen and cluster of differentiation 36 (CD36) expression was analyzed by Real-time PCR and Western blot. Lipid droplet accumulation and content was determined using Bodipy staining and enzymatic analysis. CD36 and DDR1 interaction was determined by co-immunoprecipitation. Creatinine, BUN, albuminuria, lipid content, and histological and morphological assessment of kidneys harvested from AS mice treated with Ezetimibe and/or Ramipril or vehicle was performed. Findings We demonstrate that Col I-mediated DDR1 activation induces CD36-mediated podocyte lipotoxic injury. We show that Ezetimibe interferes with the CD36/DDR1 interaction in vitro and prevents lipotoxicity in AS mice thus preserving renal function similarly to ramipril. Interpretation Our study suggests that Col I/DDR1-mediated lipotoxicity contributes to renal failure in AS and that targeting this pathway may represent a new therapeutic strategy for patients with AS and with chronic kidney diseases (CKD) associated with Col4 mutations. Funding This study is supported by the NIH grants R01DK117599, R01DK104753, R01CA227493, U54DK083912, UM1DK100846, U01DK116101, UL1TR000460 (Miami Clinical Translational Science Institute, National Center for Advancing Translational Sciences and the National Institute on Minority Health and Health Disparities), F32DK115109, Hoffmann-La Roche and Alport Syndrome Foundation.
Collapse
Affiliation(s)
- Jin-Ju Kim
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States.
| | - Judith M David
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States
| | - Sydney S Wilbon
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States
| | - Javier V Santos
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States
| | - Devang M Patel
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia
| | - Anis Ahmad
- Department of Radiation Oncology, University of Miami, FL 33136, United States
| | - Alla Mitrofanova
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States; Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Xiaochen Liu
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States
| | - Shamroop K Mallela
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States
| | - Gloria M Ducasa
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States
| | - Mengyuan Ge
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States
| | - Alexis J Sloan
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States
| | - Hassan Al-Ali
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States
| | - Marcia Boulina
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Armando J Mendez
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Gabriel N Contreras
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States
| | - Marco Prunotto
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland; School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | - Anjum Sohail
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Rafael Fridman
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Jeffrey H Miner
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Sandra Merscher
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Drug Discovery center, Department of Medicine, University of Miami Miller School of Medicine, 1580 NW 10th Ave, Miami, FL 33136, United States.
| |
Collapse
|
17
|
Jeffries DE, Borza CM, Blobaum AL, Pozzi A, Lindsley CW. Discovery of VU6015929: A Selective Discoidin Domain Receptor 1/2 (DDR1/2) Inhibitor to Explore the Role of DDR1 in Antifibrotic Therapy. ACS Med Chem Lett 2020; 11:29-33. [PMID: 31938459 DOI: 10.1021/acsmedchemlett.9b00382] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 11/25/2019] [Indexed: 01/07/2023] Open
Abstract
Herein, we report the discovery of a potent and selective dual DDR1/2 inhibitor, 7e (VU6015929), displaying low cytotoxicity, good kinome selectivity, and possessing an acceptable in vitro DMPK profile with good rodent in vivo pharmacokinetics. VU6015929 potently blocks collagen-induced DDR1 activation and collagen-IV production, suggesting DDR1 inhibition as an exciting target for antifibrotic therapy.
Collapse
Affiliation(s)
- Daniel E. Jeffries
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Corina M. Borza
- Department of Medicine, Division of Nephrology, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Anna L. Blobaum
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Ambra Pozzi
- Department of Medicine, Division of Nephrology, Vanderbilt University, Nashville, Tennessee 37232, United States
- Veterans Affairs Medical Center, Nashville, Nashville, Tennessee 37232, United States
| | - Craig W. Lindsley
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| |
Collapse
|
18
|
Salem RM, Todd JN, Sandholm N, Cole JB, Chen WM, Andrews D, Pezzolesi MG, McKeigue PM, Hiraki LT, Qiu C, Nair V, Di Liao C, Cao JJ, Valo E, Onengut-Gumuscu S, Smiles AM, McGurnaghan SJ, Haukka JK, Harjutsalo V, Brennan EP, van Zuydam N, Ahlqvist E, Doyle R, Ahluwalia TS, Lajer M, Hughes MF, Park J, Skupien J, Spiliopoulou A, Liu A, Menon R, Boustany-Kari CM, Kang HM, Nelson RG, Klein R, Klein BE, Lee KE, Gao X, Mauer M, Maestroni S, Caramori ML, de Boer IH, Miller RG, Guo J, Boright AP, Tregouet D, Gyorgy B, Snell-Bergeon JK, Maahs DM, Bull SB, Canty AJ, Palmer CNA, Stechemesser L, Paulweber B, Weitgasser R, Sokolovska J, Rovīte V, Pīrāgs V, Prakapiene E, Radzeviciene L, Verkauskiene R, Panduru NM, Groop LC, McCarthy MI, Gu HF, Möllsten A, Falhammar H, Brismar K, Martin F, Rossing P, Costacou T, Zerbini G, Marre M, Hadjadj S, McKnight AJ, Forsblom C, McKay G, Godson C, Maxwell AP, Kretzler M, Susztak K, Colhoun HM, Krolewski A, Paterson AD, Groop PH, Rich SS, Hirschhorn JN, Florez JC. Genome-Wide Association Study of Diabetic Kidney Disease Highlights Biology Involved in Glomerular Basement Membrane Collagen. J Am Soc Nephrol 2019; 30:2000-2016. [PMID: 31537649 PMCID: PMC6779358 DOI: 10.1681/asn.2019030218] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 07/08/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Although diabetic kidney disease demonstrates both familial clustering and single nucleotide polymorphism heritability, the specific genetic factors influencing risk remain largely unknown. METHODS To identify genetic variants predisposing to diabetic kidney disease, we performed genome-wide association study (GWAS) analyses. Through collaboration with the Diabetes Nephropathy Collaborative Research Initiative, we assembled a large collection of type 1 diabetes cohorts with harmonized diabetic kidney disease phenotypes. We used a spectrum of ten diabetic kidney disease definitions based on albuminuria and renal function. RESULTS Our GWAS meta-analysis included association results for up to 19,406 individuals of European descent with type 1 diabetes. We identified 16 genome-wide significant risk loci. The variant with the strongest association (rs55703767) is a common missense mutation in the collagen type IV alpha 3 chain (COL4A3) gene, which encodes a major structural component of the glomerular basement membrane (GBM). Mutations in COL4A3 are implicated in heritable nephropathies, including the progressive inherited nephropathy Alport syndrome. The rs55703767 minor allele (Asp326Tyr) is protective against several definitions of diabetic kidney disease, including albuminuria and ESKD, and demonstrated a significant association with GBM width; protective allele carriers had thinner GBM before any signs of kidney disease, and its effect was dependent on glycemia. Three other loci are in or near genes with known or suggestive involvement in this condition (BMP7) or renal biology (COLEC11 and DDR1). CONCLUSIONS The 16 diabetic kidney disease-associated loci may provide novel insights into the pathogenesis of this condition and help identify potential biologic targets for prevention and treatment.
Collapse
Affiliation(s)
- Rany M Salem
- Department of Family Medicine and Public Health, University of California San Diego, La Jolla, California
| | - Jennifer N Todd
- Division of Endocrinology, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts
- Programs in Metabolism and Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
- Center for Genomic Medicine and
| | - Niina Sandholm
- Folkhälsan Research Center, Folkhälsan Institute of Genetics, Helsinki, Finland
- Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine and
| | - Joanne B Cole
- Division of Endocrinology, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts
- Programs in Metabolism and Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
- Center for Genomic Medicine and
| | - Wei-Min Chen
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, Virginia
| | - Darrell Andrews
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - Marcus G Pezzolesi
- Division of Nephrology and Hypertension, Diabetes and Metabolism Center, University of Utah, Salt Lake City, Utah
| | - Paul M McKeigue
- Usher Institute of Population Health Sciences and Informatics and
| | - Linda T Hiraki
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Chengxiang Qiu
- Departments of Medicine and Genetics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Viji Nair
- Division of Nephrology, Department of Internal Medicine and
| | - Chen Di Liao
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jing Jing Cao
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Erkka Valo
- Folkhälsan Research Center, Folkhälsan Institute of Genetics, Helsinki, Finland
- Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine and
| | - Suna Onengut-Gumuscu
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, Virginia
| | | | - Stuart J McGurnaghan
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Jani K Haukka
- Folkhälsan Research Center, Folkhälsan Institute of Genetics, Helsinki, Finland
- Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine and
| | - Valma Harjutsalo
- Folkhälsan Research Center, Folkhälsan Institute of Genetics, Helsinki, Finland
- Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine and
- The Chronic Disease Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Eoin P Brennan
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - Natalie van Zuydam
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Emma Ahlqvist
- Department of Genomics, Diabetes and Endocrinology, Lund University Diabetes Centre, Malmö, Sweden
| | - Ross Doyle
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | | | - Maria Lajer
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Maria F Hughes
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - Jihwan Park
- Departments of Medicine and Genetics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jan Skupien
- Joslin Diabetes Center, Boston, Massachusetts
| | | | | | - Rajasree Menon
- Division of Nephrology, Department of Internal Medicine and
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | | | - Hyun M Kang
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Robert G Nelson
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona
| | - Ronald Klein
- University of Wisconsin-Madison, Madison, Wisconsin
| | | | | | - Xiaoyu Gao
- The George Washington University, Washington, DC
| | | | - Silvia Maestroni
- Complications of Diabetes Unit, Division of Immunology, Transplantation and Infectious Diseases, Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | | | | | - Rachel G Miller
- University of Pittsburgh Public Health, Pittsburgh, Pennsylvania
| | - Jingchuan Guo
- University of Pittsburgh Public Health, Pittsburgh, Pennsylvania
| | | | - David Tregouet
- INSERM UMR_S 1166, Sorbonne Université, UPMC Univ Paris 06, Paris, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, France
| | - Beata Gyorgy
- INSERM UMR_S 1166, Sorbonne Université, UPMC Univ Paris 06, Paris, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, France
| | | | - David M Maahs
- Department of Pediatrics-Endocrinology, Stanford University, Stanford, California
| | - Shelley B Bull
- The Lunenfeld-Tanenbaum Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Angelo J Canty
- Department of Mathematics and Statistics, McMaster University, Hamilton, Ontario, Canada
| | - Colin N A Palmer
- Pat Macpherson Centre for Pharmacogenetics and Pharmacogenomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Lars Stechemesser
- First Department of Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Bernhard Paulweber
- First Department of Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Raimund Weitgasser
- First Department of Medicine, Paracelsus Medical University, Salzburg, Austria
- Department of Medicine, Diakonissen-Wehrle Hospital, Salzburg, Austria
| | | | - Vita Rovīte
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Valdis Pīrāgs
- University of Latvia, Riga, Latvia
- Pauls Stradins University Hospital, Riga, Latvia
| | | | - Lina Radzeviciene
- Institute of Endocrinology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Rasa Verkauskiene
- Institute of Endocrinology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Nicolae Mircea Panduru
- Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- 2nd Clinical Department, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Leif C Groop
- Department of Genomics, Diabetes and Endocrinology, Lund University Diabetes Centre, Malmö, Sweden
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Mark I McCarthy
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, UK
- Genentech, 1 DNA Way, South San Francisco, California
| | - Harvest F Gu
- Department of Clinical Science, Intervention and Technology and
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Anna Möllsten
- Division of Pediatrics, Department of Clinical Sciences, Umeå University, Umeå, Sweden
| | - Henrik Falhammar
- Department of Molecular Medicine and Surgery, Rolf Luft Center for Diabetes Research and Endocrinology, Karolinska Institutet, Stockholm, Sweden
- Department of Endocrinology, Diabetes and Metabolism, Karolinska University Hospital, Stockholm, Sweden
| | - Kerstin Brismar
- Department of Molecular Medicine and Surgery, Rolf Luft Center for Diabetes Research and Endocrinology, Karolinska Institutet, Stockholm, Sweden
- Department of Endocrinology, Diabetes and Metabolism, Karolinska University Hospital, Stockholm, Sweden
| | - Finian Martin
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- University of Copenhagen, Copenhagen, Denmark
| | - Tina Costacou
- University of Pittsburgh Public Health, Pittsburgh, Pennsylvania
| | - Gianpaolo Zerbini
- Complications of Diabetes Unit, Division of Immunology, Transplantation and Infectious Diseases, Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Michel Marre
- Department of Diabetology, Endocrinology and Nutrition, Bichat Hospital, DHU FIRE, Assistance Publique-Hôpitaux de Paris, Paris, France
- UFR de Médecine, Paris Diderot University, Sorbonne Paris Cité, Paris, France
- INSERM UMRS 1138, Cordeliers Research Center, Paris, France
- Fondation Ophtalmologique Adolphe de Rothschild, Paris, France
| | - Samy Hadjadj
- Department of Endocrinology and Diabetology, Centre Hospitalier Universitaire de Poitiers, Poitiers, France
- INSERM CIC 1402, Poitiers, France
- L'institut du thorax, INSERM, CNRS, CHU Nantes, Nantes, France
| | - Amy J McKnight
- Centre for Public Health, Queens University of Belfast, Northern Ireland, UK
| | - Carol Forsblom
- Folkhälsan Research Center, Folkhälsan Institute of Genetics, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine and
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine and
| | - Gareth McKay
- Centre for Public Health, Queens University of Belfast, Northern Ireland, UK
| | - Catherine Godson
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - A Peter Maxwell
- Centre for Public Health, Queens University of Belfast, Northern Ireland, UK
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine and
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Katalin Susztak
- Departments of Medicine and Genetics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Helen M Colhoun
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | | | | | - Per-Henrik Groop
- Folkhälsan Research Center, Folkhälsan Institute of Genetics, Helsinki, Finland
- Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine and
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia; and
| | - Stephen S Rich
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, Virginia
| | - Joel N Hirschhorn
- Division of Endocrinology, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts
- Programs in Metabolism and Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
| | - Jose C Florez
- Programs in Metabolism and Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts;
- Center for Genomic Medicine and
- Diabetes Unit, Massachusetts General Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
19
|
Chiusa M, Hu W, Liao HJ, Su Y, Borza CM, de Caestecker MP, Skrypnyk NI, Fogo AB, Pedchenko V, Li X, Zhang MZ, Hudson BG, Basak T, Vanacore RM, Zent R, Pozzi A. The Extracellular Matrix Receptor Discoidin Domain Receptor 1 Regulates Collagen Transcription by Translocating to the Nucleus. J Am Soc Nephrol 2019; 30:1605-1624. [PMID: 31383731 PMCID: PMC6727269 DOI: 10.1681/asn.2018111160] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 05/20/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The discoidin domain receptor 1 (DDR1) is activated by collagens, upregulated in injured and fibrotic kidneys, and contributes to fibrosis by regulating extracellular matrix production, but how DDR1 controls fibrosis is poorly understood. DDR1 is a receptor tyrosine kinase (RTK). RTKs can translocate to the nucleus via a nuclear localization sequence (NLS) present on the receptor itself or a ligand it is bound to. In the nucleus, RTKs regulate gene expression by binding chromatin directly or by interacting with transcription factors. METHODS To determine whether DDR1 translocates to the nucleus and whether this event is mediated by collagen-induced DDR1 activation, we generated renal cells expressing wild-type or mutant forms of DDR1 no longer able to bind collagen. Then, we determined the location of the DDR1 upon collagen stimulation. Using both biochemical assays and immunofluorescence, we analyzed the steps involved in DDR1 nuclear translocation. RESULTS We show that although DDR1 and its natural ligand, collagen, lack an NLS, DDR1 is present in the nucleus of injured human and mouse kidney proximal tubules. We show that DDR1 nuclear translocation requires collagen-mediated receptor activation and interaction of DDR1 with SEC61B, a component of the Sec61 translocon, and nonmuscle myosin IIA and β-actin. Once in the nucleus, DDR1 binds to chromatin to increase the transcription of collagen IV, a major collagen upregulated in fibrosis. CONCLUSIONS These findings reveal a novel mechanism whereby activated DDR1 translates to the nucleus to regulate synthesis of profibrotic molecules.
Collapse
Affiliation(s)
- Manuel Chiusa
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Wen Hu
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Hong-Jun Liao
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Yan Su
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Corina M Borza
- Division of Nephrology and Hypertension, Department of Medicine, and
| | | | | | - Agnes B Fogo
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Vadim Pedchenko
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Xiyue Li
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Ming-Zhi Zhang
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Billy G Hudson
- Division of Nephrology and Hypertension, Department of Medicine, and
| | - Trayambak Basak
- Division of Nephrology and Hypertension, Department of Medicine, and
| | | | - Roy Zent
- Division of Nephrology and Hypertension, Department of Medicine, and
- Department of Veterans Affairs, Nashville, Tennessee
| | - Ambra Pozzi
- Division of Nephrology and Hypertension, Department of Medicine, and
- Department of Veterans Affairs, Nashville, Tennessee
| |
Collapse
|
20
|
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: 2.8] [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
|
21
|
Yeh YC, Lin HH, Tang MJ. Dichotomy of the function of DDR1 in cells and disease progression. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:118473. [PMID: 30954568 DOI: 10.1016/j.bbamcr.2019.04.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 12/20/2018] [Accepted: 01/06/2019] [Indexed: 12/18/2022]
Abstract
Discoidin domain receptors DDR1 and DDR2 are collagen receptor tyrosine kinases that have many roles in tissue development and disease progression. Under physiological conditions, DDR1 is predominantly expressed in epithelial cells and functions to maintain cell differentiation and tissue homeostasis. A switch in expression from DDR1 to DDR2 occurs during epithelial-to-mesenchymal transition. However, opposite effects of DDR1 are reported to be involved in the progression of cancer and fibrotic diseases. Accumulating evidence suggests that DDR1 is involved in pro-metastasis and pro-survival signals. This review summarizes the roles of DDR1 in epithelial cell differentiation, cell migration, cancer progression and tissues fibrosis and highlights how the dichotomous functions of DDR1 may relevant to different cell types and statues. Elucidation of the underlying mechanism of the dichotomous functions of DDR1 will help to develop DDR1 as a therapeutic target.
Collapse
Affiliation(s)
- Yi-Chun Yeh
- International Center for Wound Repair and Regeneration, Tainan, Taiwan
| | - Hsi-Hui Lin
- International Center for Wound Repair and Regeneration, Tainan, Taiwan; Department of Physiology, National Cheng Kung University, Tainan, Taiwan
| | - Ming-Jer Tang
- International Center for Wound Repair and Regeneration, Tainan, Taiwan; Department of Physiology, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
22
|
Moll S, Desmoulière A, Moeller MJ, Pache JC, Badi L, Arcadu F, Richter H, Satz A, Uhles S, Cavalli A, Drawnel F, Scapozza L, Prunotto M. DDR1 role in fibrosis and its pharmacological targeting. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:118474. [PMID: 30954571 DOI: 10.1016/j.bbamcr.2019.04.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 12/20/2018] [Accepted: 01/06/2019] [Indexed: 01/28/2023]
Abstract
Discoidin domain receptor1 (DDR1) is a collagen activated receptor tyrosine kinase and an attractive anti-fibrotic target. Its expression is mainly limited to epithelial cells located in several organs including skin, kidney, liver and lung. DDR1's biology is elusive, with unknown downstream activation pathways; however, it may act as a mediator of the stromal-epithelial interaction, potentially controlling the activation state of the resident quiescent fibroblasts. Increased expression of DDR1 has been documented in several types of cancer and fibrotic conditions including skin hypertrophic scars, idiopathic pulmonary fibrosis, cirrhotic liver and renal fibrosis. The present review article focuses on: a) detailing the evidence for a role of DDR1 as an anti-fibrotic target in different organs, b) clarifying DDR1 tissue distribution in healthy and diseased tissues as well as c) exploring DDR1 protective mode of action based on literature evidence and co-authors experience; d) detailing pharmacological efforts attempted to drug this subtle anti-fibrotic target to date.
Collapse
Affiliation(s)
- Solange Moll
- Department of Pathology, University Hospital of Geneva, Switzerland; Department of Pathology, Lausanne University Hospital, Switzerland
| | - Alexis Desmoulière
- Department of Physiology, Faculty of Pharmacy, University of Limoges, Limoges, France
| | - Marcus J Moeller
- Department of Nephrology and Clinical Immunology, RWTH University Hospital, Aachen, Germany
| | | | - Laura Badi
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Switzerland
| | - Filippo Arcadu
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Switzerland
| | - Hans Richter
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Switzerland
| | - Alexander Satz
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Switzerland
| | - Sabine Uhles
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Switzerland
| | - Andrea Cavalli
- Institute for Research in Biomedicine, Università della Svizzera Italiana, CH-6500, Bellinzona, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Faye Drawnel
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Switzerland
| | - Leonardo Scapozza
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | - Marco Prunotto
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Switzerland; School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
23
|
Richter H, Satz AL, Bedoucha M, Buettelmann B, Petersen AC, Harmeier A, Hermosilla R, Hochstrasser R, Burger D, Gsell B, Gasser R, Huber S, Hug MN, Kocer B, Kuhn B, Ritter M, Rudolph MG, Weibel F, Molina-David J, Kim JJ, Santos JV, Stihle M, Georges GJ, Bonfil RD, Fridman R, Uhles S, Moll S, Faul C, Fornoni A, Prunotto M. DNA-Encoded Library-Derived DDR1 Inhibitor Prevents Fibrosis and Renal Function Loss in a Genetic Mouse Model of Alport Syndrome. ACS Chem Biol 2019; 14:37-49. [PMID: 30452219 PMCID: PMC6343110 DOI: 10.1021/acschembio.8b00866] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
![]()
The
importance of Discoidin Domain Receptor 1 (DDR1) in renal fibrosis
has been shown via gene knockout and use of antisense oligonucleotides;
however, these techniques act via a reduction of DDR1 protein, while
we prove the therapeutic potential of inhibiting DDR1 phosphorylation
with a small molecule. To date, efforts to generate a selective small-molecule
to specifically modulate the activity of DDR1 in an in vivo model have been unsuccessful. We performed parallel DNA encoded
library screens against DDR1 and DDR2, and discovered a chemical series
that is highly selective for DDR1 over DDR2. Structure-guided optimization
efforts yielded the potent DDR1 inhibitor 2.45, which
possesses excellent kinome selectivity (including 64-fold selectivity
over DDR2 in a biochemical assay), a clean in vitro safety profile, and favorable pharmacokinetic and physicochemical
properties. As desired, compound 2.45 modulates DDR1
phosphorylation in vitro as well as prevents collagen-induced
activation of renal epithelial cells expressing DDR1. Compound 2.45 preserves renal function and reduces tissue damage in Col4a3–/– mice (the preclinical
mouse model of Alport syndrome) when employing a therapeutic dosing
regime, indicating the real therapeutic value of selectively inhibiting
DDR1 phosphorylation in vivo. Our results may have
wider significance as Col4a3–/– mice also represent a model for chronic kidney disease, a disease
which affects 10% of the global population.
Collapse
Affiliation(s)
- Hans Richter
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel 4070, Switzerland
| | - Alexander L. Satz
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel 4070, Switzerland
| | - Marc Bedoucha
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel 4070, Switzerland
| | - Bernd Buettelmann
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel 4070, Switzerland
| | - Ann C. Petersen
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel 4070, Switzerland
| | - Anja Harmeier
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel 4070, Switzerland
| | - Ricardo Hermosilla
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel 4070, Switzerland
| | - Remo Hochstrasser
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel 4070, Switzerland
| | - Dominique Burger
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel 4070, Switzerland
| | - Bernard Gsell
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel 4070, Switzerland
| | - Rodolfo Gasser
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel 4070, Switzerland
| | - Sylwia Huber
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel 4070, Switzerland
| | - Melanie N. Hug
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel 4070, Switzerland
| | - Buelent Kocer
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel 4070, Switzerland
| | - Bernd Kuhn
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel 4070, Switzerland
| | - Martin Ritter
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel 4070, Switzerland
| | - Markus G. Rudolph
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel 4070, Switzerland
| | - Franziska Weibel
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel 4070, Switzerland
- Ridgeline Therapeutics GmbH, Basel 4070, Switzerland
| | - Judith Molina-David
- Katz Family Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, Florida 33136, United States
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Jin-Ju Kim
- Katz Family Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, Florida 33136, United States
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Javier Varona Santos
- Katz Family Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, Florida 33136, United States
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Martine Stihle
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel 4070, Switzerland
| | - Guy J. Georges
- Roche Pharma Research and Early Development, Roche Innovation Center, Munich 82377, Germany
| | - R. Daniel Bonfil
- Department of Pathology, College of Medical Sciences, Nova Southeastern University, Fort Lauderdale, Florida 33328, United States
| | - Rafael Fridman
- Department of Pathology, Wayne State University, Detroit, Michigan 48202, United States
| | - Sabine Uhles
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel 4070, Switzerland
| | - Solange Moll
- University Hospital of Geneva, 1205 Geneva, Switzerland
| | - Christian Faul
- University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Marco Prunotto
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel 4070, Switzerland
- Office of Innovation, Immunology, Infectious Diseases & Ophthalmology (I2O), Roche and Genentech Late Stage Development, Basel 4070, Switzerland
| |
Collapse
|
24
|
New Therapies for the Treatment of Renal Fibrosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1165:625-659. [PMID: 31399988 DOI: 10.1007/978-981-13-8871-2_31] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Renal fibrosis is the common pathway for progression of chronic kidney disease (CKD) to end stage of renal disease. It is now widely accepted that the degree of renal fibrosis correlates with kidney function and CKD stages. The key cellular basis of renal fibrosis includes activation of myofibroblasts, excessive production of extracellular matrix components, and infiltration of inflammatory cells. Many cellular mechanisms responsible for renal fibrosis have been identified, and some antifibrotic agents show a greater promise in slowing down and even reversing fibrosis in animal models; however, translating basic findings into effective antifibrotic therapies in human has been limited. In this chapter, we will discuss the effects and mechanisms of some novel antifibrotic agents in both preclinical studies and clinical trials.
Collapse
|
25
|
Soomro I, Hong A, Li Z, Duncan JS, Skolnik EY. Discoidin Domain Receptor 1 (DDR1) tyrosine kinase is upregulated in PKD kidneys but does not play a role in the pathogenesis of polycystic kidney disease. PLoS One 2019; 14:e0211670. [PMID: 31260458 PMCID: PMC6602183 DOI: 10.1371/journal.pone.0211670] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 05/03/2019] [Indexed: 01/04/2023] Open
Abstract
Tolvaptan is the only drug approved to slow cyst growth and preserve kidney function in patients with autosomal dominant polycystic kidney disease (ADPKD). However, its limited efficacy combined with significant side effects underscores the need to identify new and safe therapeutic drug targets to slow progression to end stage kidney disease. We identified Discoidin Domain Receptor 1 (DDR1) as receptor tyrosine kinase upregulated in vivo in 3 mouse models of ADPKD using a novel mass spectrometry approach to identify kinases upregulated in ADPKD. Previous studies demonstrating critical roles for DDR1 to cancer progression, its potential role in the pathogenesis of a variety of other kidney disease, along with the possibility that DDR1 could provide new insight into how extracellular matrix impacts cyst growth led us to study the role of DDR1 in ADPKD pathogenesis. However, genetic deletion of DDR1 using CRISPR/Cas9 failed to slow cyst growth or preserve kidney function in both a rapid and slow mouse model of ADPKD demonstrating that DDR1 does not play a role in PKD pathogenesis and is thus a not viable drug target. In spite of the negative results, our studies will be of interest to the nephrology community as it will prevent others from potentially conducting similar experiments on DDR1 and reinforces the potential of performing unbiased screens coupled with in vivo gene editing using CRISPR/Cas9 to rapidly identify and confirm new potential drug targets for ADPKD.
Collapse
Affiliation(s)
- Irfana Soomro
- Division of Nephrology, New York University Langone Medical Center, New York, New York, United States of America
| | - Aram Hong
- Departments of Biochemistry and Molecular Pharmacology, New York University Langone Medical Center, New York, New York, United States of America
| | - Zhai Li
- Departments of Biochemistry and Molecular Pharmacology, New York University Langone Medical Center, New York, New York, United States of America
| | - James S. Duncan
- Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Edward Y. Skolnik
- Division of Nephrology, New York University Langone Medical Center, New York, New York, United States of America
- Departments of Biochemistry and Molecular Pharmacology, New York University Langone Medical Center, New York, New York, United States of America
- The Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute for Biomolecular Medicine, New York University Langone Medical Center, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
26
|
Moll S, Yasui Y, Abed A, Murata T, Shimada H, Maeda A, Fukushima N, Kanamori M, Uhles S, Badi L, Cagarelli T, Formentini I, Drawnel F, Georges G, Bergauer T, Gasser R, Bonfil RD, Fridman R, Richter H, Funk J, Moeller MJ, Chatziantoniou C, Prunotto M. Selective pharmacological inhibition of DDR1 prevents experimentally-induced glomerulonephritis in prevention and therapeutic regime. J Transl Med 2018; 16:148. [PMID: 29859097 PMCID: PMC5984769 DOI: 10.1186/s12967-018-1524-5] [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: 04/30/2018] [Accepted: 05/23/2018] [Indexed: 11/10/2022] Open
Abstract
Background Discoidin domain receptor 1 (DDR1) is a collagen-activated receptor tyrosine kinase extensively implicated in diseases such as cancer, atherosclerosis and fibrosis. Multiple preclinical studies, performed using either a gene deletion or a gene silencing approaches, have shown this receptor being a major driver target of fibrosis and glomerulosclerosis. Methods The present study investigated the role and relevance of DDR1 in human crescentic glomerulonephritis (GN). Detailed DDR1 expression was first characterized in detail in human GN biopsies using a novel selective anti-DDR1 antibody using immunohistochemistry. Subsequently the protective role of DDR1 was investigated using a highly selective, novel, small molecule inhibitor in a nephrotoxic serum (NTS) GN model in a prophylactic regime and in the NEP25 GN mouse model using a therapeutic intervention regime. Results DDR1 expression was shown to be mainly limited to renal epithelium. In humans, DDR1 is highly induced in injured podocytes, in bridging cells expressing both parietal epithelial cell (PEC) and podocyte markers and in a subset of PECs forming the cellular crescents in human GN. Pharmacological inhibition of DDR1 in NTS improved both renal function and histological parameters. These results, obtained using a prophylactic regime, were confirmed in the NEP25 GN mouse model using a therapeutic intervention regime. Gene expression analysis of NTS showed that pharmacological blockade of DDR1 specifically reverted fibrotic and inflammatory gene networks and modulated expression of the glomerular cell gene signature, further validating DDR1 as a major mediator of cell fate in podocytes and PECs. Conclusions Together, these results suggest that DDR1 inhibition might be an attractive and promising pharmacological intervention for the treatment of GN, predominantly by targeting the renal epithelium. Electronic supplementary material The online version of this article (10.1186/s12967-018-1524-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Solange Moll
- Department of Pathology, University Hospital of Geneva, Geneva, Switzerland
| | - Yukari Yasui
- Research Division, Chugai Pharmaceutical Co., Ltd, Tokyo, Japan
| | - Ahmed Abed
- INSERM, UMR S 1155, Hôpital Tenon, 75020, Paris, France
| | - Takeshi Murata
- Research Division, Chugai Pharmaceutical Co., Ltd, Tokyo, Japan
| | - Hideaki Shimada
- Research Division, Chugai Pharmaceutical Co., Ltd, Tokyo, Japan.,Chugai Pharmabody Research Pte. Ltd., Singapore, Singapore
| | - Akira Maeda
- Research Division, Chugai Pharmaceutical Co., Ltd, Tokyo, Japan
| | | | - Masakazu Kanamori
- Research Division, Chugai Pharmaceutical Co., Ltd, Tokyo, Japan.,Chugai Pharmabody Research Pte. Ltd., Singapore, Singapore
| | - Sabine Uhles
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Laura Badi
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Thomas Cagarelli
- Department of Pathology, University Hospital of Geneva, Geneva, Switzerland
| | - Ivan Formentini
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland.,Late Stage, AstraZeneca, Göteborgs, Sweden
| | - Faye Drawnel
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Guy Georges
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Munich, Germany
| | - Tobias Bergauer
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Rodolfo Gasser
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - R Daniel Bonfil
- Department of Pathology, College of Medical Sciences, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Rafael Fridman
- Department of Pathology, Wayne State University, Detroit, MI, USA
| | - Hans Richter
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Juergen Funk
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Marcus J Moeller
- Department of Nephrology and Clinical Immunology, RWTH University, Aachen, Germany
| | | | - Marco Prunotto
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland. .,Office of Innovation, Immunology, Infectious Diseases & Ophthalmology (I2O), Roche and Genentech Late Stage Development, 124 Grenzacherstrasse, 4070, Basel, Switzerland. .,School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
27
|
Coelho NM, McCulloch CA. Mechanical signaling through the discoidin domain receptor 1 plays a central role in tissue fibrosis. Cell Adh Migr 2018; 12:348-362. [PMID: 29513135 PMCID: PMC6363045 DOI: 10.1080/19336918.2018.1448353] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/20/2018] [Accepted: 02/26/2018] [Indexed: 02/08/2023] Open
Abstract
The preservation of tissue and organ architecture and function depends on tightly regulated interactions of cells with the extracellular matrix (ECM). These interactions are maintained in a dynamic equilibrium that balances intracellular, myosin-generated tension with extracellular resistance conferred by the mechanical properties of the extracellular matrix. Disturbances of this equilibrium can lead to the development of fibrotic lesions that are associated with a wide repertoire of high prevalence diseases including obstructive cardiovascular diseases, muscular dystrophy and cancer. Mechanotransduction is the process by which mechanical cues are converted into biochemical signals. At the core of mechanotransduction are sensory systems, which are frequently located at sites of cell-ECM and cell-cell contacts. As integrins (cell-ECM junctions) and cadherins (cell-cell contacts) have been extensively studied, we focus here on the properties of the discoidin domain receptor 1 (DDR1), a tyrosine kinase that mediates cell adhesion to collagen. DDR1 expression is positively associated with fibrotic lesions of heart, kidney, liver, lung and perivascular tissues. As the most common end-point of all fibrotic disorders is dysregulated collagen remodeling, we consider here the mechanical signaling functions of DDR1 in processing of fibrillar collagen that lead to tissue fibrosis.
Collapse
Affiliation(s)
- Nuno M. Coelho
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
28
|
Abstract
Discoidin Domain Receptor 1 (DDR1) belongs to a family of two non-integrin collagen receptors, DDR1 and DDR2, which display a tyrosine kinase activity. DDR1 has been widely studied in different kind of pathologies including chronic kidney diseases (CKD). The aims of this commentary are 1. to review the existing information about DDR1 expression in healthy and diseased kidney, 2. to comment the data highlighting DDR1 as a major actor in CKD, 3. to suggest areas of research which require further investigation to better characterize the signaling pathways regulating DDR1 role in CKD. The results recapitulated in this commentary emphasize the involvement of DDR1 in the pro-inflammatory and pro-fibrotic processes which drives the development of CKD. They also underline the beneficial effect of its blockade in pre-clinical models and thus, reinforce its status of interesting therapeutic target.
Collapse
Affiliation(s)
- Aude Dorison
- a Inserm UMR S 1155 and Sorbonne Université , Paris , France
| | | |
Collapse
|
29
|
Ma TKW, McAdoo SP, Tam FWK. Targeting the tyrosine kinase signalling pathways for treatment of immune-mediated glomerulonephritis: from bench to bedside and beyond. Nephrol Dial Transplant 2017; 32:i129-i138. [PMID: 28391340 PMCID: PMC5410974 DOI: 10.1093/ndt/gfw336] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 08/18/2016] [Indexed: 12/25/2022] Open
Abstract
Glomerulonephritis (GN) affects patients of all ages and is an important cause of morbidity and mortality. Non-selective immunosuppressive drugs have been used in immune-mediated GN but often result in systemic side effects and occasionally fatal infective complications. There is increasing evidence from both preclinical and clinical studies that abnormal activation of receptor and non-receptor tyrosine kinase signalling pathways are implicated in the pathogenesis of immune-mediated GN. Activation of spleen tyrosine kinase (SYK), Bruton's tyrosine kinase (BTK), platelet-derived growth factor receptor (PDGFR), epidermal growth factor receptor (EGFR) and discoidin domain receptor 1 (DDR1) have been demonstrated in anti-GBM disease. SYK is implicated in the pathogenesis of ANCA-associated GN. SYK, BTK, PDGFR, EFGR, DDR1 and Janus kinase are implicated in the pathogenesis of lupus nephritis. A representative animal model of IgA nephropathy (IgAN) is lacking. Based on the results from in vitro and human renal biopsy study results, a phase II clinical trial is ongoing to evaluate the efficacy and safety of fostamatinib (an oral SYK inhibitor) in high-risk IgAN patient. Various tyrosine kinase inhibitors (TKIs) have been approved for cancer treatment. Clinical trials of TKIs in GN may be justified given their long-term safety data. In this review we will discuss the current unmet medical needs in GN treatment and research as well as the current stage of development of TKIs in GN treatment and propose an accelerated translational research approach to investigate whether selective inhibition of tyrosine kinase provides a safer and more efficacious option for GN treatment.
Collapse
Affiliation(s)
- Terry King-Wing Ma
- Renal and Vascular Inflammation Section, Department of Medicine, Imperial College London, Hammersmith Hospital, Du Cane Road, London, UK.,Carol and Richard Yu Peritoneal Dialysis Research Centre, Department of Medicine and Therapeutics, Prince of Wales Hospital, Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Stephen P McAdoo
- Renal and Vascular Inflammation Section, Department of Medicine, Imperial College London, Hammersmith Hospital, Du Cane Road, London, UK
| | - Frederick Wai Keung Tam
- Renal and Vascular Inflammation Section, Department of Medicine, Imperial College London, Hammersmith Hospital, Du Cane Road, London, UK
| |
Collapse
|
30
|
Aguilera KY, Huang H, Du W, Hagopian MM, Wang Z, Hinz S, Hwang TH, Wang H, Fleming JB, Castrillon DH, Ren X, Ding K, Brekken RA. Inhibition of Discoidin Domain Receptor 1 Reduces Collagen-mediated Tumorigenicity in Pancreatic Ductal Adenocarcinoma. Mol Cancer Ther 2017; 16:2473-2485. [PMID: 28864681 DOI: 10.1158/1535-7163.mct-16-0834] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 05/04/2017] [Accepted: 07/28/2017] [Indexed: 12/14/2022]
Abstract
The extracellular matrix (ECM), a principal component of pancreatic ductal adenocarcinoma (PDA), is rich in fibrillar collagens that facilitate tumor cell survival and chemoresistance. Discoidin domain receptor 1 (DDR1) is a receptor tyrosine kinase that specifically binds fibrillar collagens and has been implicated in promoting cell proliferation, migration, adhesion, ECM remodeling, and response to growth factors. We found that collagen-induced activation of DDR1 stimulated protumorigenic signaling through protein tyrosine kinase 2 (PYK2) and pseudopodium-enriched atypical kinase 1 (PEAK1) in pancreatic cancer cells. Pharmacologic inhibition of DDR1 with an ATP-competitive orally available small-molecule kinase inhibitor (7rh) abrogated collagen-induced DDR1 signaling in pancreatic tumor cells and consequently reduced colony formation and migration. Furthermore, the inhibition of DDR1 with 7rh showed striking efficacy in combination with chemotherapy in orthotopic xenografts and autochthonous pancreatic tumors where it significantly reduced DDR1 activation and downstream signaling, reduced primary tumor burden, and improved chemoresponse. These data demonstrate that targeting collagen signaling in conjunction with conventional cytotoxic chemotherapy has the potential to improve outcome for pancreatic cancer patients. Mol Cancer Ther; 16(11); 2473-85. ©2017 AACR.
Collapse
Affiliation(s)
- Kristina Y Aguilera
- Division of Surgical Oncology, Department of Surgery and Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas
| | - Huocong Huang
- Division of Surgical Oncology, Department of Surgery and Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas
| | - Wenting Du
- Division of Surgical Oncology, Department of Surgery and Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas
| | - Moriah M Hagopian
- Division of Surgical Oncology, Department of Surgery and Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas
| | - Zhen Wang
- School of Pharmacy, Jinan University, Guangzhou, China
| | - Stefan Hinz
- Division of Surgical Oncology, Department of Surgery and Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas
| | - Tae Hyun Hwang
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Huamin Wang
- Department of Pathology, UT MD Anderson Cancer Center, Houston, Texas
| | - Jason B Fleming
- Department of Surgical Oncology, UT MD Anderson Cancer Center, Houston, Texas
| | - Diego H Castrillon
- Department of Clinical Science, UT Southwestern Medical Center, Dallas, Texas
| | - Xiaomei Ren
- School of Pharmacy, Jinan University, Guangzhou, China
| | - Ke Ding
- School of Pharmacy, Jinan University, Guangzhou, China
| | - Rolf A Brekken
- Division of Surgical Oncology, Department of Surgery and Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas. .,Department of Pharmacology, UT Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
31
|
Kavvadas P, Abed A, Poulain C, Authier F, Labéjof LP, Calmont A, Afieri C, Prakoura N, Dussaule JC, Chatziantoniou C, Chadjichristos CE. Decreased Expression of Connexin 43 Blunts the Progression of Experimental GN. J Am Soc Nephrol 2017; 28:2915-2930. [PMID: 28667079 DOI: 10.1681/asn.2016111211] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 05/05/2017] [Indexed: 11/03/2022] Open
Abstract
GN refers to a variety of renal pathologies that often progress to ESRD, but the molecular mechanisms underlying this progression remain incompletely characterized. Here, we determined whether dysregulated expression of the gap junction protein connexin 43, which has been observed in the progression of renal disease, contributes to GN progression. Immunostaining revealed de novo expression of connexin 43 in damaged glomeruli in patients with glomerular diseases as well as in mice after induction of experimental GN. Notably, 2 weeks after the induction of GN with nephrotoxic serum, mice with a heterozygous deletion of the connexin 43 gene (connexin 43+/-) had proteinuria, BUN, and serum creatinine levels significantly lower than those of wild-type animals. Additionally, the connexin 43+/- mice showed less crescent formation, tubular dilation, monocyte infiltration, and interstitial renal fibrosis. Treatment of cultured podocytes with connexin 43-specific blocking peptides attenuated TGF-β-induced cytoskeletal and morphologic changes and apoptosis as did treatment with the purinergic blocker suramin. Finally, therapeutic treatment of GN mice with connexin 43-specific antisense oligodeoxynucleotide improved functional and structural renal parameters. These findings suggest that crosstalk between connexin 43 and purinergic signaling contributes to podocyte damage in GN. Given that this protein is highly induced in individuals with glomerular diseases, connexin 43 may be a novel target for therapeutic treatment of GN.
Collapse
Affiliation(s)
- Panagiotis Kavvadas
- National Institute for Health and Medical Research Unité Mixte de Recherche-S1155, Batiment Recherche, Tenon Hospital, Paris, France
| | - Ahmed Abed
- National Institute for Health and Medical Research Unité Mixte de Recherche-S1155, Batiment Recherche, Tenon Hospital, Paris, France.,Sorbonne Universites, University Pierre et Marie Curie University Paris 6, Paris, France
| | - Coralie Poulain
- National Institute for Health and Medical Research Unité Mixte de Recherche-S1155, Batiment Recherche, Tenon Hospital, Paris, France.,University René Descartes, Paris, France.,University Denis Diderot, Paris, France
| | - Florence Authier
- National Institute for Health and Medical Research Unité Mixte de Recherche-S1155, Batiment Recherche, Tenon Hospital, Paris, France
| | - Lise-Paule Labéjof
- National Institute for Health and Medical Research Unité Mixte de Recherche-S1155, Batiment Recherche, Tenon Hospital, Paris, France.,Universidade Estadual de Santa Cruz, Ilhéus, Bahia, Brazil
| | - Amelie Calmont
- National Institute for Health and Medical Research Unité Mixte de Recherche-S1155, Batiment Recherche, Tenon Hospital, Paris, France
| | - Carlo Afieri
- National Institute for Health and Medical Research Unité Mixte de Recherche-S1155, Batiment Recherche, Tenon Hospital, Paris, France.,Unit of Nephrology Dialysis and Kidney Transplantation, Fondazione Istituto Di Ricovero e Cura a Carattere Scientifico Ca Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy; and
| | - Niki Prakoura
- National Institute for Health and Medical Research Unité Mixte de Recherche-S1155, Batiment Recherche, Tenon Hospital, Paris, France
| | - Jean-Claude Dussaule
- National Institute for Health and Medical Research Unité Mixte de Recherche-S1155, Batiment Recherche, Tenon Hospital, Paris, France.,Sorbonne Universites, University Pierre et Marie Curie University Paris 6, Paris, France.,Department of Physiology, Saint Antoine Hospital, Paris, France
| | - Christos Chatziantoniou
- National Institute for Health and Medical Research Unité Mixte de Recherche-S1155, Batiment Recherche, Tenon Hospital, Paris, France.,Sorbonne Universites, University Pierre et Marie Curie University Paris 6, Paris, France
| | - Christos E Chadjichristos
- National Institute for Health and Medical Research Unité Mixte de Recherche-S1155, Batiment Recherche, Tenon Hospital, Paris, France; .,Sorbonne Universites, University Pierre et Marie Curie University Paris 6, Paris, France
| |
Collapse
|
32
|
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: 1.7] [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
|
33
|
Plasma heme-induced renal toxicity is related to a capillary rarefaction. Sci Rep 2017; 7:40156. [PMID: 28071761 PMCID: PMC5223203 DOI: 10.1038/srep40156] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 12/02/2016] [Indexed: 01/07/2023] Open
Abstract
Severe hypertension can lead to malignant hypertension (MH) with renal thrombotic microangiopathy and hemolysis. The role of plasma heme release in this setting is unknown. We aimed at evaluating the effect of a mild plasma heme increase by hemin administration in angiotensin II (AngII)-mediated hypertensive rats. Prevalence of MH and blood pressure values were similar in AngII and AngII + hemin groups. MH rats displayed a decreased renal blood flow (RBF), increased renal vascular resistances (RVR), and increased aorta and interlobar arteries remodeling with a severe renal microcirculation assessed by peritubular capillaries (PTC) rarefaction. Hemin-treated rats with or without AngII displayed also a decreased RBF and increased RVR explained only by PCT rarefaction. In AngII rats, RBF was similar to controls (with increased RVR). PTC density appeared strongly correlated to tubular damage score (rho = -0.65, p < 0.0001) and also renal Heme Oygenase-1 (HO-1) mRNA (rho = -0.67, p < 0.0001). HO-1 was expressed in PTC and renal tubules in MH rats, but only in PTC in other groups. In conclusion, though increased plasma heme does not play a role in triggering or aggravating MH, heme release appears as a relevant toxic mediator leading to renal impairment, primarily through PTC endothelial dysfunction rather than direct tubular toxicity.
Collapse
|
34
|
Borza CM, Su Y, Tran TL, Yu L, Steyns N, Temple KJ, Skwark MJ, Meiler J, Lindsley CW, Hicks BR, Leitinger B, Zent R, Pozzi A. Discoidin domain receptor 1 kinase activity is required for regulating collagen IV synthesis. Matrix Biol 2017; 57-58:258-271. [PMID: 27915093 PMCID: PMC5329129 DOI: 10.1016/j.matbio.2016.11.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 11/21/2016] [Accepted: 11/22/2016] [Indexed: 01/29/2023]
Abstract
Discoidin domain receptor 1 (DDR1) is a receptor tyrosine kinase that binds to and is activated by collagens. DDR1 expression increases following kidney injury and accumulating evidence suggests that it contributes to the progression of injury. To this end, deletion of DDR1 is beneficial in ameliorating kidney injury induced by angiotensin infusion, unilateral ureteral obstruction, or nephrotoxic nephritis. Most of the beneficial effects observed in the DDR1-null mice are attributed to reduced inflammatory cell infiltration to the site of injury, suggesting that DDR1 plays a pro-inflammatory effect. The goal of this study was to determine whether, in addition to its pro-inflammatory effect, DDR1 plays a deleterious effect in kidney injury by directly regulating extracellular matrix production. We show that DDR1-null mice have reduced deposition of glomerular collagens I and IV as well as decreased proteinuria following the partial renal ablation model of kidney injury. Using mesangial cells isolated from DDR1-null mice, we show that these cells produce significantly less collagen compared to DDR1-null cells reconstituted with wild type DDR1. Moreover, mutagenesis analysis revealed that mutations in the collagen binding site or in the kinase domain significantly reduce DDR1-mediated collagen production. Finally, we provide evidence that blocking DDR1 kinase activity with an ATP-competitive small molecule inhibitor reduces collagen production. In conclusion, our studies indicate that the kinase activity of DDR1 plays a key role in DDR1-induced collagen synthesis and suggest that blocking collagen-mediated DDR1 activation may be beneficial in fibrotic diseases.
Collapse
Affiliation(s)
- Corina M Borza
- Department of Medicine (Division of Nephrology), Vanderbilt University, Nashville, TN, United States
| | - Yan Su
- Department of Medicine (Division of Nephrology), Vanderbilt University, Nashville, TN, United States
| | - Truc-Linh Tran
- Department of Medicine (Division of Nephrology), Vanderbilt University, Nashville, TN, United States
| | - Ling Yu
- Department of Medicine (Division of Nephrology), Vanderbilt University, Nashville, TN, United States
| | - Nick Steyns
- Department Chemistry, Pharmacology, Vanderbilt University, Nashville, TN, United States
| | - Kayla J Temple
- Department Chemistry, Pharmacology, Vanderbilt University, Nashville, TN, United States
| | - Marcin J Skwark
- Department Chemistry, Pharmacology, Vanderbilt University, Nashville, TN, United States
| | - Jens Meiler
- Department Chemistry, Pharmacology, Vanderbilt University, Nashville, TN, United States
| | - Craig W Lindsley
- Department Chemistry, Pharmacology, Vanderbilt University, Nashville, TN, United States
| | - Brennan R Hicks
- Department of Medicine (Division of Nephrology), Vanderbilt University, Nashville, TN, United States
| | - Birgit Leitinger
- National Heart and Lung Institute, Imperial College of London, London, UK
| | - Roy Zent
- Department of Medicine (Division of Nephrology), Vanderbilt University, Nashville, TN, United States; Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States; Veterans Affairs Hospitals, Nashville, TN, United States
| | - Ambra Pozzi
- Department of Medicine (Division of Nephrology), Vanderbilt University, Nashville, TN, United States; Veterans Affairs Hospitals, Nashville, TN, United States.
| |
Collapse
|
35
|
Contribution of collagen adhesion receptors to tissue fibrosis. Cell Tissue Res 2016; 365:521-38. [DOI: 10.1007/s00441-016-2440-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 06/01/2016] [Indexed: 02/07/2023]
|
36
|
Liu F, Zhuang S. Role of Receptor Tyrosine Kinase Signaling in Renal Fibrosis. Int J Mol Sci 2016; 17:ijms17060972. [PMID: 27331812 PMCID: PMC4926504 DOI: 10.3390/ijms17060972] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 05/23/2016] [Accepted: 06/01/2016] [Indexed: 12/31/2022] Open
Abstract
Renal fibrosis can be induced in different renal diseases, but ultimately progresses to end stage renal disease. Although the pathophysiologic process of renal fibrosis have not been fully elucidated, it is characterized by glomerulosclerosis and/or tubular interstitial fibrosis, and is believed to be caused by the proliferation of renal inherent cells, including glomerular epithelial cells, mesangial cells, and endothelial cells, along with defective kidney repair, renal interstitial fibroblasts activation, and extracellular matrix deposition. Receptor tyrosine kinases (RTKs) regulate a variety of cell physiological processes, including metabolism, growth, differentiation, and survival. Many studies from in vitro and animal models have provided evidence that RTKs play important roles in the pathogenic process of renal fibrosis. It is also showed that tyrosine kinases inhibitors (TKIs) have anti-fibrotic effects in basic research and clinical trials. In this review, we summarize the evidence for involvement of specific RTKs in renal fibrosis process and the employment of TKIs as a therapeutic approach for renal fibrosis.
Collapse
Affiliation(s)
- Feng Liu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
- Department of Medicine, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA.
| |
Collapse
|
37
|
Kerroch M, Alfieri C, Dorison A, Boffa JJ, Chatziantoniou C, Dussaule JC. Protective effects of genetic inhibition of Discoidin Domain Receptor 1 in experimental renal disease. Sci Rep 2016; 6:21262. [PMID: 26880216 PMCID: PMC4754689 DOI: 10.1038/srep21262] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 01/20/2016] [Indexed: 11/09/2022] Open
Abstract
Chronic kidney disease is a progressive incurable pathology affecting millions of people. Intensive investigations aim to identify targets for therapy. We have previously demonstrated that abnormal expression of the Discoidin Domain Receptor 1 (DDR1) is a key factor of renal disease by promoting inflammation and fibrosis. The present study investigates whether blocking the expression of DDR1 after the initiation of renal disease can delay or arrest the progression of this pathology. Severe renal disease was induced by either injecting nephrotoxic serum (NTS) or performing unilateral ureteral obstruction in mice, and the expression of DDR1 was inhibited by administering antisense oligodeoxynucleotides either at 4 or 8 days after NTS (corresponding to early or more established phases of disease, respectively), or at day 2 after ligation. DDR1 antisense administration at day 4 stopped the increase of proteinuria and protected animals against the progression of glomeruloneprhitis, as evidenced by functional, structural and cellular indexes. Antisense administration at day 8 delayed progression -but to a smaller degree- of renal disease. Similar beneficial effects on renal structure and inflammation were observed with the antisense administration of DDR1 after ureteral ligation. Thus, targeting DDR1 can be a promising strategy in the treatment of chronic kidney disease.
Collapse
Affiliation(s)
- Monique Kerroch
- INSERM UMR S 1155, Hôpital Tenon, 75020 Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France
| | - Carlo Alfieri
- INSERM UMR S 1155, Hôpital Tenon, 75020 Paris, France.,Department of Medicine and Medical Specialties, Unit of Nephrology, Dialysis, and Renal Transplant, Fondazione Istituto di Ricerca e Cura a Carattere Scientifico Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Aude Dorison
- INSERM UMR S 1155, Hôpital Tenon, 75020 Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France
| | - Jean-Jacques Boffa
- INSERM UMR S 1155, Hôpital Tenon, 75020 Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,Service de Néphrologie et Dialyses, Hôpital Tenon, AP-HP, Paris, France
| | - Christos Chatziantoniou
- INSERM UMR S 1155, Hôpital Tenon, 75020 Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France
| | - Jean-Claude Dussaule
- INSERM UMR S 1155, Hôpital Tenon, 75020 Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,Department of Physiology, Saint-Antoine Hospital, AP-HP, Paris, France
| |
Collapse
|
38
|
Marchal PO, Kavvadas P, Abed A, Kazazian C, Authier F, Koseki H, Hiraoka S, Boffa JJ, Martinerie C, Chadjichristos CE. Reduced NOV/CCN3 Expression Limits Inflammation and Interstitial Renal Fibrosis after Obstructive Nephropathy in Mice. PLoS One 2015; 10:e0137876. [PMID: 26367310 PMCID: PMC4569074 DOI: 10.1371/journal.pone.0137876] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 08/23/2015] [Indexed: 11/20/2022] Open
Abstract
The main hallmark of chronic kidney disease (CKD) is excessive inflammation leading to interstitial tissue fibrosis. It has been recently reported that NOV/CCN3 could be involved in kidney damage but its role in the progression of nephropathies is poorly known. NOV/CCN3 is a secreted multifunctional protein belonging to the CCN family involved in different physiological and pathological processes such as angiogenesis, inflammation and cancers. The purpose of our study was to determine the role of NOV/CCN3 in renal inflammation and fibrosis related to primitive tubulointerstitial injury. After unilateral ureteral obstruction (UUO), renal histology and real-time PCR were performed in NOV/CCN3-/- and wild type mice. NOV/CCN3 mRNA expression was increased in the obstructed kidneys in the early stages of the obstructive nephropathy. Interestingly, plasmatic levels of NOV/CCN3 were strongly induced after 7 days of UUO and the injection of recombinant NOV/CCN3 protein in healthy mice significantly increased CCL2 mRNA levels. Furthermore, after 7 days of UUO NOV/CCN3-/- mice displayed reduced proinflammatory cytokines and adhesion markers expression leading to restricted accumulation of interstitial monocytes, in comparison with their wild type littermates. Consequently, in NOV/CCN3-/- mice interstitial renal fibrosis was blunted after 15 days of UUO. In agreement with our experimental data, NOV/CCN3 expression was highly increased in biopsies of patients with tubulointerstitial nephritis. Thus, the inhibition of NOV/CCN3 may represent a novel target for the progression of renal diseases.
Collapse
Affiliation(s)
- Pierre-Olivier Marchal
- INSERM, UMR-S938, Centre de Recherche Saint-Antoine, Saint-Antoine Hospital, Paris, France
- INSERM UMR-S1155, Tenon Hospital, Paris, France
- Sorbonne Universités, UPMC Univ Paris 6, Paris, France
| | | | - Ahmed Abed
- INSERM UMR-S1155, Tenon Hospital, Paris, France
- Sorbonne Universités, UPMC Univ Paris 6, Paris, France
| | - Chantal Kazazian
- INSERM, UMR-S938, Centre de Recherche Saint-Antoine, Saint-Antoine Hospital, Paris, France
| | | | - Haruhiko Koseki
- RIKEN Research Center for Allergy and Immunology (RCAI), RIKEN Yokohama Institute, Yokohama, Japan
| | - Shuichi Hiraoka
- Department of Biochemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Jean-Jacques Boffa
- INSERM UMR-S1155, Tenon Hospital, Paris, France
- Department of Nephrology, Tenon Hospital, Paris, France
| | - Cécile Martinerie
- INSERM, UMR-S938, Centre de Recherche Saint-Antoine, Saint-Antoine Hospital, Paris, France
- Sorbonne Universités, UPMC Univ Paris 6, Paris, France
| | - Christos E. Chadjichristos
- INSERM UMR-S1155, Tenon Hospital, Paris, France
- Sorbonne Universités, UPMC Univ Paris 6, Paris, France
- * E-mail:
| |
Collapse
|
39
|
Delestré L, Bakey Z, Prado C, Hoffmann S, Bihoreau MT, Lelongt B, Gauguier D. ANKS3 Co-Localises with ANKS6 in Mouse Renal Cilia and Is Associated with Vasopressin Signaling and Apoptosis In Vivo in Mice. PLoS One 2015; 10:e0136781. [PMID: 26327442 PMCID: PMC4556665 DOI: 10.1371/journal.pone.0136781] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 08/07/2015] [Indexed: 02/07/2023] Open
Abstract
Mutations in Ankyrin repeat and sterile alpha motif domain containing 6 (ANKS6) play a causative role in renal cyst formation in the PKD/Mhm(cy/+) rat model of polycystic kidney disease and in nephronophthisis in humans. A network of protein partners of ANKS6 is emerging and their functional characterization provides important clues to understand the role of ANKS6 in renal biology and in mechanisms involved in the formation of renal cysts. Following experimental confirmation of interaction between ANKS6and ANKS3 using a Yeast two hybrid system, we demonstrated that binding between the two proteins occurs through their sterile alpha motif (SAM) and that the amino acid 823 in rat ANSK6 is key for this interaction. We further showed their interaction by co-immunoprecipitation and showed in vivo in mice that ANKS3 is present in renal cilia. Downregulated expression of Anks3 in vivo in mice by Locked Nucleic Acid (LNA) modified antisense oligonucleotides was associated with increased transcription of vasopressin-induced genes, suggesting changes in renal water permeability, and altered transcription of genes encoding proteins involved in cilium structure, apoptosis and cell proliferation. These data provide experimental evidence of ANKS3-ANKS6 direct interaction through their SAM domain and co-localisation in mouse renal cilia, and shed light on molecular mechanisms indirectly mediated by ANKS6 in the mouse kidney, that may be affected by altered ANKS3-ANKS6 interaction. Our results contribute to improved knowledge of the structure and function of the network of proteins interacting with ANKS6, which may represent therapeutic targets in cystic diseases.
Collapse
Affiliation(s)
- Laure Delestré
- Sorbonne Universities, University Pierre and Marie Curie, University Paris Descartes, Sorbonne Paris Cité, INSERM, UMR_S1138, Cordeliers Research Centre, Paris, France
| | - Zeineb Bakey
- Sorbonne Universities, University Pierre and Marie Curie, UMR_S1155, Paris, France
- INSERM, UMR_S1155 Hôpital Tenon, Paris, France
| | - Cécilia Prado
- Sorbonne Universities, University Pierre and Marie Curie, University Paris Descartes, Sorbonne Paris Cité, INSERM, UMR_S1138, Cordeliers Research Centre, Paris, France
| | - Sigrid Hoffmann
- Medical Research Centre, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | | | - Brigitte Lelongt
- Sorbonne Universities, University Pierre and Marie Curie, UMR_S1155, Paris, France
- INSERM, UMR_S1155 Hôpital Tenon, Paris, France
| | - Dominique Gauguier
- Sorbonne Universities, University Pierre and Marie Curie, University Paris Descartes, Sorbonne Paris Cité, INSERM, UMR_S1138, Cordeliers Research Centre, Paris, France
- Institute of Cardiometabolism & Nutrition, Pitié-Salpêtrière Hospital, University Pierre and Marie-Curie, Paris, France
- * E-mail:
| |
Collapse
|
40
|
Chen Z, Migeon T, Verpont MC, Zaidan M, Sado Y, Kerjaschki D, Ronco P, Plaisier E. HANAC Syndrome Col4a1 Mutation Causes Neonate Glomerular Hyperpermeability and Adult Glomerulocystic Kidney Disease. J Am Soc Nephrol 2015; 27:1042-54. [PMID: 26260163 DOI: 10.1681/asn.2014121217] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 06/16/2015] [Indexed: 12/21/2022] Open
Abstract
Hereditary angiopathy, nephropathy, aneurysms, and muscle cramps (HANAC) syndrome is an autosomal dominant syndrome caused by mutations in COL4A1 that encodes the α1 chain of collagen IV, a major component of basement membranes. Patients present with cerebral small vessel disease, retinal tortuosity, muscle cramps, and kidney disease consisting of multiple renal cysts, chronic kidney failure, and sometimes hematuria. Mutations producing HANAC syndrome localize within the integrin binding site containing CB3[IV] fragment of the COL4A1 protein. To investigate the pathophysiology of HANAC syndrome, we generated mice harboring the Col4a1 p.Gly498Val mutation identified in a family with the syndrome. Col4a1 G498V mutation resulted in delayed glomerulogenesis and podocyte differentiation without reduction of nephron number, causing albuminuria and hematuria in newborns. The glomerular defects resolved within the first month, but glomerular cysts developed in 3-month-old mutant mice. Abnormal structure of Bowman's capsule was associated with metalloproteinase induction and activation of the glomerular parietal epithelial cells that abnormally expressed CD44,α-SMA, ILK, and DDR1. Inflammatory infiltrates were observed around glomeruli and arterioles. Homozygous Col4a1 G498V mutant mice additionally showed dysmorphic papillae and urinary concentration defects. These results reveal a developmental role for the α1α1α2 collagen IV molecule in the embryonic glomerular basement membrane, affecting podocyte differentiation. The observed association between molecular alteration of the collagenous network in Bowman's capsule of the mature kidney and activation of parietal epithelial cells, matrix remodeling, and inflammation may account for glomerular cyst development and CKD in patients with COL4A1-related disorders.
Collapse
Affiliation(s)
- Zhiyong Chen
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche (UMR)S 1155, Paris, France
| | - Tiffany Migeon
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche (UMR)S 1155, Paris, France; Sorbonne University, Université Pierre et Marie Curie, Paris 06, UMR_S 1155, Paris, France
| | - Marie-Christine Verpont
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche (UMR)S 1155, Paris, France; Sorbonne University, Université Pierre et Marie Curie, Paris 06, UMR_S 1155, Paris, France
| | - Mohamad Zaidan
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche (UMR)S 1155, Paris, France
| | - Yoshikazu Sado
- Division of Immunology, Shigei Medical Research Institute, Okayama, Japan
| | - Dontscho Kerjaschki
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria; and
| | - Pierre Ronco
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche (UMR)S 1155, Paris, France; Sorbonne University, Université Pierre et Marie Curie, Paris 06, UMR_S 1155, Paris, France; Assistance Publique-Hôpitaux de Paris, Department of Nephrology and Dialysis, Tenon Hospital, Paris, France
| | - Emmanuelle Plaisier
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche (UMR)S 1155, Paris, France; Sorbonne University, Université Pierre et Marie Curie, Paris 06, UMR_S 1155, Paris, France; Assistance Publique-Hôpitaux de Paris, Department of Nephrology and Dialysis, Tenon Hospital, Paris, France
| |
Collapse
|
41
|
Alfieri C, Kavvadas P, Simonini P, Ikehata M, Dussaule JC, Chadjichristos CE, Rastaldi MP, Messa P, Chatziantoniou C. Discoidin domain receptor-1 and periostin: new players in chronic kidney disease. Nephrol Dial Transplant 2015; 30:1965-71. [PMID: 25829327 DOI: 10.1093/ndt/gfv074] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 02/05/2015] [Indexed: 11/14/2022] Open
Abstract
The incidence and prevalence of chronic kidney disease represents an important problem for public health. In renal diseases, the main histologic alterations derive from the development of renal fibrosis which results from the loss of the balance between pro- and anti-fibrotic factors. Tyrosine kinase receptors (RTKs) and matricellular proteins (MPs) are nowadays studied as potential modulators of renal injury. RTKs regulate cell cycle, migration, metabolism and cellular differentiation. Discoidin domain receptor-1 (DDR-1) is an RTK that has been extensively studied in cancer, and lung and renal diseases. It modulates inflammatory recruitment, extracellular matrix deposition and fibrosis; in renal diseases, it appears to act independently of the underlying disease. MPs regulate cell-matrix interactions and matrix accumulation, cellular adhesion and migration, and expression of inflammatory cells. Periostin is an MP, mainly studied in bone, heart, lung and cancer. Several studies demonstrated that it mediates cell-matrix interactions, migration of inflammatory cells and development of fibrosis. Recently, it has been reported in several nephropathies. In this review, we discuss the potential pathological roles of DDR-1 and periostin focussing on the kidney in both experimental models and human diseases.
Collapse
Affiliation(s)
- Carlo Alfieri
- Institut National de la Santé et de la Recherche Médicale Research Unit S_1155, Bâtiment Recherche, Tenon Hospital, Paris, France Department of Medicine and Medical Specialties, Unit of Nephrology, Dialysis, and Renal Transplant, Fondazione Istituto di Ricerca e Cura a Carattere Scientifico Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Panagiotis Kavvadas
- Institut National de la Santé et de la Recherche Médicale Research Unit S_1155, Bâtiment Recherche, Tenon Hospital, Paris, France
| | - Paola Simonini
- Department of Medicine and Medical Specialties, Unit of Nephrology, Dialysis, and Renal Transplant, Fondazione Istituto di Ricerca e Cura a Carattere Scientifico Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Masami Ikehata
- Research Laboratory of Nephrology, Fondazione Istituto di Ricerca e Cura a Carattere Scientifico Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Jean Claude Dussaule
- Institut National de la Santé et de la Recherche Médicale Research Unit S_1155, Bâtiment Recherche, Tenon Hospital, Paris, France
| | - Christos E Chadjichristos
- Institut National de la Santé et de la Recherche Médicale Research Unit S_1155, Bâtiment Recherche, Tenon Hospital, Paris, France
| | - Maria Pia Rastaldi
- Research Laboratory of Nephrology, Fondazione Istituto di Ricerca e Cura a Carattere Scientifico Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Piergiorgio Messa
- Department of Medicine and Medical Specialties, Unit of Nephrology, Dialysis, and Renal Transplant, Fondazione Istituto di Ricerca e Cura a Carattere Scientifico Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Christos Chatziantoniou
- Institut National de la Santé et de la Recherche Médicale Research Unit S_1155, Bâtiment Recherche, Tenon Hospital, Paris, France
| |
Collapse
|
42
|
Li Y, Lu X, Ren X, Ding K. Small Molecule Discoidin Domain Receptor Kinase Inhibitors and Potential Medical Applications. J Med Chem 2015; 58:3287-301. [DOI: 10.1021/jm5012319] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Yupeng Li
- State Key Laboratory
of Respiratory Diseases, Guangzhou Institutes of Biomedicine and Health,
Chinese Academy of Sciences, No. 190
Kaiyuan Avenue, Guangzhou 510530, China
- University of Chinese Academy of Sciences, No. 19 Yuquan Road, Beijing 100049, China
| | - Xiaoyun Lu
- State Key Laboratory
of Respiratory Diseases, Guangzhou Institutes of Biomedicine and Health,
Chinese Academy of Sciences, No. 190
Kaiyuan Avenue, Guangzhou 510530, China
| | - Xiaomei Ren
- State Key Laboratory
of Respiratory Diseases, Guangzhou Institutes of Biomedicine and Health,
Chinese Academy of Sciences, No. 190
Kaiyuan Avenue, Guangzhou 510530, China
| | - Ke Ding
- State Key Laboratory
of Respiratory Diseases, Guangzhou Institutes of Biomedicine and Health,
Chinese Academy of Sciences, No. 190
Kaiyuan Avenue, Guangzhou 510530, China
| |
Collapse
|
43
|
Borza CM, Chen X, Zent R, Pozzi A. Cell Receptor-Basement Membrane Interactions in Health and Disease: A Kidney-Centric View. CURRENT TOPICS IN MEMBRANES 2015; 76:231-53. [PMID: 26610916 PMCID: PMC4913201 DOI: 10.1016/bs.ctm.2015.07.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cell-extracellular matrix (ECM) interactions are essential for tissue development, homeostasis, and response to injury. Basement membranes (BMs) are specialized ECMs that separate epithelial or endothelial cells from stromal components and interact with cells via cellular receptors, including integrins and discoidin domain receptors. Disruption of cell-BM interactions due to either injury or genetic defects in either the ECM components or cellular receptors often lead to irreversible tissue injury and loss of organ function. Animal models that lack specific BM components or receptors either globally or in selective tissues have been used to help with our understanding of the molecular mechanisms whereby cell-BM interactions regulate organ function in physiological and pathological conditions. We review recently published works on animal models that explore how cell-BM interactions regulate kidney homeostasis in both health and disease.
Collapse
Affiliation(s)
- Corina M. Borza
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, 37232
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - Xiwu Chen
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - Roy Zent
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, 37232
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN, 37232
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN, 37232
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, 37232
- Department of Medicine, Veterans Administration Hospital, Nashville, TN, 37232
| | - Ambra Pozzi
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, 37232
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN, 37232
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN, 37232
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, 37232
- Department of Medicine, Veterans Administration Hospital, Nashville, TN, 37232
| |
Collapse
|
44
|
Leitinger B. Discoidin domain receptor functions in physiological and pathological conditions. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 310:39-87. [PMID: 24725424 DOI: 10.1016/b978-0-12-800180-6.00002-5] [Citation(s) in RCA: 253] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The discoidin domain receptors, DDR1 and DDR2, are nonintegrin collagen receptors that are members of the receptor tyrosine kinase family. Both DDRs bind a number of different collagen types and play important roles in embryo development. Dysregulated DDR function is associated with progression of various human diseases, including fibrosis, arthritis, and cancer. By interacting with key components of the extracellular matrix and displaying distinct activation kinetics, the DDRs form a unique subfamily of receptor tyrosine kinases. DDR-facilitated cellular functions include cell migration, cell survival, proliferation, and differentiation, as well as remodeling of extracellular matrices. This review summarizes the current knowledge of DDR-ligand interactions, DDR-initiated signal pathways and the molecular mechanisms that regulate receptor function. Also discussed are the roles of DDRs in development and disease progression.
Collapse
Affiliation(s)
- Birgit Leitinger
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.
| |
Collapse
|
45
|
Abstract
Over the last decade, identification and characterization of novel markers of progression and targets for therapy of chronic kidney disease (CKD) have been challenging for the research community. Several promising candidates have emerged, mainly from experimental models of CKD that are yet to be investigated in clinical studies. The authors identified two candidate genes: periostin, an extracellular matrix protein involved in bone and dental development, and the discoidin domain receptor 1 (DDR1), a collagen-binding membrane receptor with tyrosine kinase activity. Both genes are inactive in adulthood under normal conditions but have been shown to be highly inducible following injury to glomerular or tubular epithelial cells. The objective of this review is to summarize recent evidence supporting the role of periostin and DDR1 as potential novel biomarkers and therapeutic targets in CKD.
Collapse
|
46
|
Moll S, Meier M, Formentini I, Pomposiello S, Prunotto M. New renal drug development to face chronic renal disease. Expert Opin Drug Discov 2014; 9:1471-85. [DOI: 10.1517/17460441.2014.956075] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
47
|
Reiser J, Sever S, Faul C. Signal transduction in podocytes--spotlight on receptor tyrosine kinases. Nat Rev Nephrol 2014; 10:104-15. [PMID: 24394191 PMCID: PMC4109315 DOI: 10.1038/nrneph.2013.274] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The mammalian kidney filtration barrier is a complex multicellular, multicomponent structure that maintains homeostasis by regulating electrolytes, acid-base balance, and blood pressure (via maintenance of salt and water balance). To perform these multiple functions, podocytes--an important component of the filtration apparatus--must process a series of intercellular signals. Integrating these signals with diverse cellular responses enables a coordinated response to various conditions. Although mature podocytes are terminally differentiated and cannot proliferate, they are able to respond to growth factors. It is possible that the initial response of podocytes to growth factors is beneficial and protective, and might include the induction of hypertrophic cell growth. However, extended and/or uncontrolled growth factor signalling might be maladaptive and could result in the induction of apoptosis and podocyte loss. Growth factors signal via the activation of receptor tyrosine kinases (RTKs) on their target cells and around a quarter of the 58 RTK family members that are encoded in the human genome have been identified in podocytes. Pharmacological inhibitors of many RTKs exist and are currently used in experimental and clinical cancer therapy. The identification of pathological RTK-mediated signal transduction pathways in podocytes could provide a starting point for the development of novel therapies for glomerular disorders.
Collapse
Affiliation(s)
- Jochen Reiser
- Department of Medicine, Rush University Medical Center, 1735 West Harrison Street, Cohn Building, Suite 724, Chicago, IL 60612, USA
| | - Sanja Sever
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, 149 13th Street, Charlestown, MA 02129, USA
| | - Christian Faul
- Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, 1580 North West 10th Avenue (R-762), Batchelor Building 626, Miami, FL 33136, USA
| |
Collapse
|
48
|
Discoidin domain receptors in disease. Matrix Biol 2013; 34:185-92. [PMID: 24361528 DOI: 10.1016/j.matbio.2013.12.002] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 12/05/2013] [Accepted: 12/05/2013] [Indexed: 12/17/2022]
Abstract
Discoidin domain receptors, DDR1 and DDR2, lie at the intersection of two large receptor families, namely the extracellular matrix and tyrosine kinase receptors. As such, DDRs are uniquely positioned to function as sensors for extracellular matrix and to regulate a wide range of cell functions from migration and proliferation to cytokine secretion and extracellular matrix homeostasis/remodeling. While activation of DDRs by extracellular matrix collagens is required for normal development and tissue homeostasis, aberrant activation of these receptors following injury or in disease is detrimental. The availability of mice lacking DDRs has enabled us to identify key roles played by these receptors in disease initiation and progression. DDR1 promotes inflammation in atherosclerosis, lung fibrosis and kidney injury, while DDR2 contributes to osteoarthritis. Furthermore, both DDRs have been implicated in cancer progression. Yet the mechanisms whereby DDRs contribute to disease progression are poorly understood. In this review we highlight the mechanisms whereby DDRs regulate two important processes, namely inflammation and tissue fibrosis. In addition, we discuss the challenges of targeting DDRs in disease. Selective targeting of these receptors requires understanding of how they interact with and are activated by extracellular matrix, and whether their cellular function is dependent on or independent of receptor kinase activity.
Collapse
|
49
|
Abstract
Snapshot of key developments in the patent literature accompanied by explanatory synopses
Collapse
|
50
|
Everett JR. Report on the 10th anniversary of international drug discovery science and technology conference, 8 - 10 november 2012, nanjing, china. Expert Opin Drug Discov 2013; 8:357-61. [PMID: 23339328 DOI: 10.1517/17460441.2013.762353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The 10th Anniversary of International Drug Discovery Science and Technology (IDDST) Conference was held in Nanjing, China from 8 to 10 November 2012. The conference ran in parallel with the 2nd Annual Symposium of Drug Delivery Systems. Over 400 delegates from both conferences came together for the Opening Ceremony and Keynote Addresses but otherwise pursued separate paths in the huge facilities of the Nanjing International Expo Centre. The IDDST was arranged into 19 separate Chapters covering drug discovery biology, target validation, chemistry, rational drug design, pharmacology and toxicology, drug screening technology, 'omics' technologies, analytical, automation and enabling technologies, informatics, stem cells and regenerative medicine, bioprocessing, generics, biosimilars and biologicals and seven disease areas: cancer, CNS, respiratory and inflammation, autoimmune, emerging infectious, bone and orphan diseases. There were also two sessions of a 'Bench to Bedside to Business' Program and a Chinese Scientist programme. In each period of the IDDST conference, up to seven sessions were running in parallel. This Meeting Highlight samples just a fraction of the content of this large meeting. The talks included have as a link, the use of new approaches to drug discovery. Many other excellent talks could have been highlighted and the author has necessarily had to be selective.
Collapse
Affiliation(s)
- Jeremy R Everett
- University of Greenwich, School of Science, Chatham Maritime, Kent, UK.
| |
Collapse
|