1
|
Grillo E, Ravelli C, Colleluori G, D'Agostino F, Domenichini M, Giordano A, Mitola S. Role of gremlin-1 in the pathophysiology of the adipose tissues. Cytokine Growth Factor Rev 2023; 69:51-60. [PMID: 36155165 DOI: 10.1016/j.cytogfr.2022.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/12/2022] [Indexed: 02/07/2023]
Abstract
Gremlin-1 is a secreted bone morphogenetic protein (BMP) antagonist playing a pivotal role in the regulation of tissue formation and embryonic development. Since its first identification in 1997, gremlin-1 has been shown to be a multifunctional factor involved in wound healing, inflammation, cancer and tissue fibrosis. Among others, the activity of gremlin-1 is mediated by its interaction with BMPs or with membrane receptors such as the vascular endothelial growth factor receptor 2 (VEGFR2) or heparan sulfate proteoglycans (HSPGs). Growing evidence has highlighted a central role of gremlin-1 in the homeostasis of the adipose tissue (AT). Of note, gremlin-1 is involved in AT dysfunction during type 2 diabetes, obesity and non-alcoholic fatty liver disease (NAFLD) metabolic disorders. In this review we discuss recent findings on gremlin-1 involvement in AT biology, with particular attention to its role in metabolic diseases, to highlight its potential as a prognostic marker and therapeutic target.
Collapse
Affiliation(s)
- Elisabetta Grillo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| | - Cosetta Ravelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Georgia Colleluori
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Via Tronto 10/A, 60020 Ancona, Italy
| | - Francesco D'Agostino
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Mattia Domenichini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Antonio Giordano
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Via Tronto 10/A, 60020 Ancona, Italy
| | - Stefania Mitola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
2
|
Pérez-Lozano ML, Sudre L, van Eegher S, Citadelle D, Pigenet A, Lafage-Proust MH, Pastoureau P, De Ceuninck F, Berenbaum F, Houard X. Gremlin-1 and BMP-4 Overexpressed in Osteoarthritis Drive an Osteochondral-Remodeling Program in Osteoblasts and Hypertrophic Chondrocytes. Int J Mol Sci 2022; 23:ijms23042084. [PMID: 35216203 PMCID: PMC8874623 DOI: 10.3390/ijms23042084] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 02/04/2023] Open
Abstract
Osteoarthritis (OA) is a whole joint disease characterized by an important remodeling of the osteochondral junction. It includes cartilage mineralization due to chondrocyte hypertrophic differentiation and bone sclerosis. Here, we investigated whether gremlin-1 (Grem-1) and its BMP partners could be involved in the remodeling events of the osteochondral junction in OA. We found that Grem-1, BMP-2, and BMP-4 immunostaining was detected in chondrocytes from the deep layer of cartilage and in subchondral bone of knee OA patients, and was positively correlated with cartilage damage. ELISA assays showed that bone released more Grem-1 and BMP-4 than cartilage, which released more BMP-2. In vitro experiments evidenced that compression stimulated the expression and the release of Grem-1 and BMP-4 by osteoblasts. Grem-1 was also overexpressed during the prehypertrophic to hypertrophic differentiation of murine articular chondrocytes. Recombinant Grem-1 stimulated Mmp-3 and Mmp-13 expression in murine chondrocytes and osteoblasts, whereas recombinant BMP-4 stimulated the expression of genes associated with angiogenesis (Angptl4 and osteoclastogenesis (Rankl and Ccl2). In conclusion, Grem-1 and BMP-4, whose expression at the osteochondral junction increased with OA progression, may favor the pathological remodeling of the osteochondral junction by inducing a catabolic and tissue remodeling program in hypertrophic chondrocytes and osteoblasts.
Collapse
Affiliation(s)
- Maria-Luisa Pérez-Lozano
- Centre de Recherche Saint-Antoine (CRSA), INSERM, Sorbonne Université, F-75012 Paris, France; (M.-L.P.-L.); (L.S.); (S.v.E.); (D.C.); (A.P.); (X.H.)
| | - Laure Sudre
- Centre de Recherche Saint-Antoine (CRSA), INSERM, Sorbonne Université, F-75012 Paris, France; (M.-L.P.-L.); (L.S.); (S.v.E.); (D.C.); (A.P.); (X.H.)
| | - Sandy van Eegher
- Centre de Recherche Saint-Antoine (CRSA), INSERM, Sorbonne Université, F-75012 Paris, France; (M.-L.P.-L.); (L.S.); (S.v.E.); (D.C.); (A.P.); (X.H.)
| | - Danièle Citadelle
- Centre de Recherche Saint-Antoine (CRSA), INSERM, Sorbonne Université, F-75012 Paris, France; (M.-L.P.-L.); (L.S.); (S.v.E.); (D.C.); (A.P.); (X.H.)
| | - Audrey Pigenet
- Centre de Recherche Saint-Antoine (CRSA), INSERM, Sorbonne Université, F-75012 Paris, France; (M.-L.P.-L.); (L.S.); (S.v.E.); (D.C.); (A.P.); (X.H.)
| | | | - Philippe Pastoureau
- Immuno-Inflammatory Diseases Department, Servier Research Institute, F-92150 Suresnes, France; (P.P.); (F.D.C.)
| | - Frédéric De Ceuninck
- Immuno-Inflammatory Diseases Department, Servier Research Institute, F-92150 Suresnes, France; (P.P.); (F.D.C.)
| | - Francis Berenbaum
- Centre de Recherche Saint-Antoine (CRSA), INSERM, Sorbonne Université, F-75012 Paris, France; (M.-L.P.-L.); (L.S.); (S.v.E.); (D.C.); (A.P.); (X.H.)
- Centre de Recherche Saint-Antoine (CRSA), INSERM, Sorbonne Université, AP-HP Hôpital Saint Antoine, F-75012 Paris, France
- Correspondence:
| | - Xavier Houard
- Centre de Recherche Saint-Antoine (CRSA), INSERM, Sorbonne Université, F-75012 Paris, France; (M.-L.P.-L.); (L.S.); (S.v.E.); (D.C.); (A.P.); (X.H.)
| |
Collapse
|
3
|
Elemam NM, Malek AI, Mahmoud EE, El-Huneidi W, Talaat IM. Insights into the Role of Gremlin-1, a Bone Morphogenic Protein Antagonist, in Cancer Initiation and Progression. Biomedicines 2022; 10:301. [PMID: 35203511 PMCID: PMC8869528 DOI: 10.3390/biomedicines10020301] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 02/07/2023] Open
Abstract
The bone morphogenic protein (BMP) antagonist Gremlin-1 is a biologically significant regulator known for its crucial role in tissue differentiation and embryonic development. Nevertheless, it has been reported that Gremlin-1 can exhibit its function through BMP dependent and independent pathways. Gremlin-1 has also been reported to be involved in organ fibrosis, which has been correlated to the development of other diseases, such as renal inflammation and diabetic nephropathy. Based on growing evidence, Gremlin-1 has recently been implicated in the initiation and progression of different types of cancers. Further, it contributes to the stemness state of cancer cells. Herein, we explore the recent findings on the role of Gremlin-1 in various cancer types, including breast, cervical, colorectal, and gastric cancers, as well as glioblastomas. Additionally, we highlighted the impact of Gremlin-1 on cellular processes and signaling pathways involved in carcinogenesis. Therefore, it was suggested that Gremlin-1 might be a promising prognostic biomarker and therapeutic target in cancers.
Collapse
Affiliation(s)
- Noha Mousaad Elemam
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates; (N.M.E.); (A.I.M.); (E.E.M.)
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Abdullah Imadeddin Malek
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates; (N.M.E.); (A.I.M.); (E.E.M.)
| | - Esraa Elaraby Mahmoud
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates; (N.M.E.); (A.I.M.); (E.E.M.)
| | - Waseem El-Huneidi
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates; (N.M.E.); (A.I.M.); (E.E.M.)
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Iman M. Talaat
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates; (N.M.E.); (A.I.M.); (E.E.M.)
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Faculty of Medicine, Alexandria University, Alexandria 21526, Egypt
| |
Collapse
|
4
|
Marquez-Exposito L, Cantero-Navarro E, R Rodrigues-Diez R, Orejudo M, Tejera-Muñoz A, Tejedor L, Rayego-Mateos S, Rández-Carbayo J, Santos-Sanchez L, Mezzano S, Lavoz C, Ruiz-Ortega M. Molecular Regulation of Notch Signaling by Gremlin. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1227:81-94. [PMID: 32072500 DOI: 10.1007/978-3-030-36422-9_6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Gremlin is a member of the TGF-β superfamily that can act as a BMP antagonist, and recently, has been described as a ligand of the vascular endothelial growth factor receptor 2 (VEGFR2). Gremlin shares properties with the Notch signaling pathway. Both participate in embryonic development and are reactivated in pathological conditions. Gremlin is emerging as a potential therapeutic target and biomarker of renal diseases. Here we review the role of the Gremlin-VEGFR2 axis in renal damage and downstream signaling mechanisms, such as Notch pathway.
Collapse
Affiliation(s)
- Laura Marquez-Exposito
- Cellular and Molecular Biology in Renal and Vascular Pathology, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain.,Red de Investigación Renal (REDINREN), Madrid, Spain
| | - Elena Cantero-Navarro
- Cellular and Molecular Biology in Renal and Vascular Pathology, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain.,Red de Investigación Renal (REDINREN), Madrid, Spain
| | - Raúl R Rodrigues-Diez
- Cellular and Molecular Biology in Renal and Vascular Pathology, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain.,Red de Investigación Renal (REDINREN), Madrid, Spain
| | - Macarena Orejudo
- Cellular and Molecular Biology in Renal and Vascular Pathology, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain.,Red de Investigación Renal (REDINREN), Madrid, Spain
| | - Antonio Tejera-Muñoz
- Cellular and Molecular Biology in Renal and Vascular Pathology, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain.,Red de Investigación Renal (REDINREN), Madrid, Spain
| | - Lucia Tejedor
- Cellular and Molecular Biology in Renal and Vascular Pathology, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain.,Red de Investigación Renal (REDINREN), Madrid, Spain
| | - Sandra Rayego-Mateos
- Cellular and Molecular Biology in Renal and Vascular Pathology, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain.,Vascular and Renal Translational Research Group, Institut de Recerca Biomèdica de Lleida IRBLleida, Lleida, Spain
| | - Javier Rández-Carbayo
- Cellular and Molecular Biology in Renal and Vascular Pathology, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain.,Red de Investigación Renal (REDINREN), Madrid, Spain
| | - Laura Santos-Sanchez
- Cellular and Molecular Biology in Renal and Vascular Pathology, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain.,Red de Investigación Renal (REDINREN), Madrid, Spain
| | - Sergio Mezzano
- Division of Nephrology, School of Medicine, Universidad Austral, Valdivia, Chile
| | - Carolina Lavoz
- Division of Nephrology, School of Medicine, Universidad Austral, Valdivia, Chile
| | - Marta Ruiz-Ortega
- Cellular and Molecular Biology in Renal and Vascular Pathology, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain. .,Red de Investigación Renal (REDINREN), Madrid, Spain.
| |
Collapse
|
5
|
Gu Q, Luo Y, Chen C, Jiang D, Huang Q, Wang X. GREM1 overexpression inhibits proliferation, migration and angiogenesis of osteosarcoma. Exp Cell Res 2019; 384:111619. [PMID: 31525341 DOI: 10.1016/j.yexcr.2019.111619] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 08/31/2019] [Accepted: 09/06/2019] [Indexed: 12/12/2022]
Abstract
Osteosarcoma is the most common malignancy of bone that occurs in young adults and children, with a five-year survival rate of 60-70%. Metastasis of osteosarcoma maintains an even poorer prognosis. GREM1 plays an important role in regulating organogenesis, body patterning, and tissue differentiation. However, there are limited studies on GREM1 in osteosarcomas. This study was carried out to characterize the expression and function of GREM1 in osteosarcoma cells, thus extending our understanding of osteosarcoma metastasis. GREM1 expression was detected in hBMSC, hFOB1.19, Saos-2, MG63 and U2OS cell lines using quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot analysis. Gain- and loss-of-function approaches were used to assess the biological function of GREM1 in U2OS cells. The effects of GREM1 on U2OS cell proliferation were examined using the CCK-8 and colony formation assay. Migration and invasion ability were confirmed by the wound healing and Transwell assay, respectively. Flow cytometry was used to analyse the effect of GREM1 on the cell cycle and apoptosis. The expression of GREM1 targets was evaluated by qRT-PCR and western blotting. The expression of GREM1 was significantly downregulated in osteosarcoma. GREM1 overexpression inhibited the proliferation, migration and invasion of U2OS cells. GREM1 overexpression suppressed tumour cell-induced endothelial cell migration and invasion ability. The effect of GREM1 may be transduced through regulation of the BMP target transcription factor inhibitor of MMP-2 and -9 as well as Id1. GREM1 overexpression and knockdown regulates the tumorigenesis of osteosarcoma in vivo. In conclusion, GREM1 is downregulated in osteosarcoma cells, and overexpression of GREM1 inhibits the proliferation, migration, invasion and angiogenesis abilities of osteosarcoma cells in vitro and in vivo.
Collapse
Affiliation(s)
- Qingguo Gu
- Department of Orthopedic Oncology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Yibin Luo
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Cheng Chen
- Department of Orthopedics, Shanghai University of Medicine &health Sciences Affiliated Zhoupu Hospital, China
| | - Dongjie Jiang
- Department of Orthopedic Oncology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China.
| | - Quan Huang
- Department of Orthopedic Oncology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China.
| | - Xinwei Wang
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China.
| |
Collapse
|
6
|
Mezzano S, Droguett A, Lavoz C, Krall P, Egido J, Ruiz-Ortega M. Gremlin and renal diseases: ready to jump the fence to clinical utility? Nephrol Dial Transplant 2019; 33:735-741. [PMID: 28992340 DOI: 10.1093/ndt/gfx194] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 04/18/2017] [Indexed: 12/31/2022] Open
Abstract
The current therapeutic strategy for the treatment of chronic kidney diseases only ameliorates disease progression. During renal injury, developmental genes are re-expressed and could be potential therapeutic targets. Among those genes reactivated in the adult damaged kidney, Gremlin is of particular relevance since recent data suggest that it could be a mediator of diabetic nephropathy and other progressive renal diseases. Earlier studies have shown that Gremlin is upregulated in trans-differentiated renal proximal tubular cells and in several chronic kidney diseases associated with fibrosis. However, not much was known about the mechanisms by which Gremlin acts in renal pathophysiology. The role of Gremlin as a bone morphogenetic protein antagonist has clearly been demonstrated in organogenesis and in fibrotic-related disorders. Gremlin binds to vascular endothelial growth factor receptor 2 (VEGFR2) in endothelial and tubular epithelial cells. Activation of the Gremlin-VEGFR2 axis was found in several human nephropathies. We have recently described that Gremlin activates the VEGFR2 signaling pathway in the kidney, eliciting a downstream mechanism linked to renal inflammatory response. Gremlin deletion improves experimental renal damage, diminishing fibrosis. Overall, the available data identify the Gremlin-VEGFR2 axis as a novel therapeutic target for kidney inflammation and fibrosis and provide a rationale for unveiling new concepts to investigate in several clinical conditions.
Collapse
Affiliation(s)
- Sergio Mezzano
- Division of Nephrology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Alejandra Droguett
- Division of Nephrology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Carolina Lavoz
- Division of Nephrology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Paola Krall
- Division of Nephrology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Jesús Egido
- Division of Nephrology and Hypertension, University Hospital, Fundación Jiménez Díaz-Universidad Autónoma, CIBERDEM, Instituto Renal Reina Sofía, Madrid, Spain
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, Universidad Autónoma Madrid, IIS-Fundación Jiménez Díaz, RedinRen, Madrid, Spain
| |
Collapse
|
7
|
Exercise Attenuates Acute β-Adrenergic Overactivation-Induced Cardiac Fibrosis by Modulating Cytokines. J Cardiovasc Transl Res 2019; 12:528-538. [PMID: 31161536 DOI: 10.1007/s12265-019-09894-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 05/13/2019] [Indexed: 10/26/2022]
Abstract
During acute sympathetic stress, the overactivation of β-adrenergic receptors (β-ARs) causes cardiac fibrosis by triggering inflammation and cytokine expression. It is unknown whether exercise training inhibits acute β-AR overactivation-induced cytokine expression and cardiac injury. Here, we report that running exercise inhibited cardiac fibrosis and improved cardiac function in mice treated with isoproterenol (ISO), a β-AR agonist. A cytokine antibody array revealed that running exercise prevented most of the changes in cytokine expression induced by ISO. Specifically, ISO-induced upregulation of 18 cytokines was prevented by running exercise. A Kyoto encyclopedia of genes and genomes analysis of these cytokines revealed that Hedgehog and RAP1 signaling pathways were involved in the regulation of cytokine expression by exercise. The changes in the expression of some cytokines that were prevented by exercise were verified by an enzyme-linked immunosorbent assay and real-time PCR. In conclusion, running exercise prevented the cytokine expression changes after acute β-AR overactivation and therefore attenuated cardiac fibrosis. Acute sympathetic stress is an important risk factor for the patients with cardiovascular diseases, and the present study revealed that exercise training can prevent against the upregulation of cytokines and the subsequent cardiac injury induced by acute sympathetic stress, suggesting that exercise training may be beneficial for cardiovascular patients who are in risk of acute sympathetic stress. This finding provides a theoretical basis for the application of exercise training in patients who may suffer from acute sympathetic stress.
Collapse
|
8
|
Droguett A, Valderrama G, Burgos ME, Carpio D, Saka C, Egido J, Ruiz-Ortega M, Mezzano S. Gremlin, A Potential Urinary Biomarker of Anca-Associated Crescentic Glomerulonephritis. Sci Rep 2019; 9:6867. [PMID: 31053735 PMCID: PMC6499786 DOI: 10.1038/s41598-019-43358-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 04/23/2019] [Indexed: 01/08/2023] Open
Abstract
Gremlin renal overexpression has been reported in diabetic nephropathy, pauci-immune crescentic glomerulonephritis and chronic allograft nephropathy and has been implicated in the pathophysiology of the progression of renal damage. However, it is unknown whether urinary Gremlin can be associated with renal functional status, renal biopsy findings and outcome. To examine these associations we studied 20 patients with ANCA+ renal vasculitis and very high urinary Gremlin (354 ± 76 ug/gCr), 86 patients with other glomerular diseases and moderately elevated urinary Gremlin (83 ± 14 ug/gCr) and 11 healthy controls (urinary Gremlin 11.3 ± 2.4 ug/gCr). Urinary Gremlin was significantly correlated with renal expression of Gremlin (r = 0.64, p = 0.013) observed in cellular glomerular crescents, tubular epithelial cells and interstitial inflammatory cells. Moreover, urinary Gremlin levels were correlated with the number of glomerular crescents (r = 0.53; p < 0.001), renal CD68 positive cells (r = 0.71; p < 0.005), tubulointerstitial fibrosis (r = 0.50; p < 0.05), and serum creatinine levels (r = 0.60; p < 0.001). Interestingly, Gremlin expression was colocalized with CD68, CD163 (monocyte/macrophage markers) and CCL18 positive cells. ROC curve analysis showed that the cutoff value of urinary Gremlin in glomerular diseases as 43 ug/gCr with 72% of sensitivity and 100% of specificity [AUC: 0.96 (CI 95% 0.92–0.99] (p < 0.001). For ANCA+ renal vasculitis the value of urinary Gremlin of 241 ug/gCr had 55% of sensitivity and 100% of specificity [AUC: 0.81 (CI 95% 0.68–0.94) (p < 0.001]. Based on these results we propose that urinary Gremlin represents a non-invasive biomarker in ANCA+ renal vasculitis, and suggest a role of Gremlin in the formation of crescents.
Collapse
Affiliation(s)
- Alejandra Droguett
- Nephrology Division, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Graciela Valderrama
- Nephrology Division, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - María E Burgos
- Nephrology Division, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Daniel Carpio
- Nephrology Division, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Constanza Saka
- Nephrology Division, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Jesús Egido
- Cellular Biology in Renal Disease Laboratory, Universidad Autónoma. IIS-Fundación Jiménez Díaz, Madrid, Spain
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Disease Laboratory, Universidad Autónoma. IIS-Fundación Jiménez Díaz, Madrid, Spain
| | - Sergio Mezzano
- Nephrology Division, School of Medicine, Universidad Austral de Chile, Valdivia, Chile.
| |
Collapse
|
9
|
Excessive mechanical loading promotes osteoarthritis through the gremlin-1-NF-κB pathway. Nat Commun 2019; 10:1442. [PMID: 30926814 PMCID: PMC6441020 DOI: 10.1038/s41467-019-09491-5] [Citation(s) in RCA: 207] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 03/14/2019] [Indexed: 12/22/2022] Open
Abstract
Exposure of articular cartilage to excessive mechanical loading is deeply involved in the pathogenesis of osteoarthritis. Here, we identify gremlin-1 as a mechanical loading-inducible factor in chondrocytes, detected at high levels in middle and deep layers of cartilage after cyclic strain or hydrostatic pressure loading. Gremlin-1 activates nuclear factor-κB signalling, leading to subsequent induction of catabolic enzymes. In mice intra-articular administration of gremlin-1 antibody or chondrocyte-specific deletion of Gremlin-1 decelerates osteoarthritis development, while intra-articular administration of recombinant gremlin-1 exacerbates this process. Furthermore, ras-related C3 botulinum toxin substrate 1 activation induced by mechanical loading enhances reactive oxygen species (ROS) production. Amongst ROS-activating transcription factors, RelA/p65 induces Gremlin-1 transcription, which antagonizes induction of anabolic genes such as Sox9, Col2a1, and Acan by bone morphogenetic proteins. Thus, gremlin-1 plays essential roles in cartilage degeneration by excessive mechanical loading. Excessive mechanical stress promotes the development of osteoarthritis. Here Chang et al. identify gremlin-1 as a factor expressed in chondrocytes in response to mechanical stress, and contributing to osteoarthritis via activation of the NF-κB pathway.
Collapse
|
10
|
Marquez-Exposito L, Lavoz C, Rodrigues-Diez RR, Rayego-Mateos S, Orejudo M, Cantero-Navarro E, Ortiz A, Egido J, Selgas R, Mezzano S, Ruiz-Ortega M. Gremlin Regulates Tubular Epithelial to Mesenchymal Transition via VEGFR2: Potential Role in Renal Fibrosis. Front Pharmacol 2018; 9:1195. [PMID: 30386246 PMCID: PMC6199372 DOI: 10.3389/fphar.2018.01195] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 09/28/2018] [Indexed: 12/27/2022] Open
Abstract
Chronic kidney disease (CKD) is emerging as an important health problem due to the increase number of CKD patients and the absence of an effective curative treatment. Gremlin has been proposed as a novel therapeutic target for renal inflammatory diseases, acting via Vascular Endothelial Growth Factor Receptor-2 (VEGFR2). Although many evidences suggest that Gremlin could regulate renal fibrosis, the receptor involved has not been yet clarified. Gremlin, as other TGF-β superfamily members, regulates tubular epithelial to mesenchymal transition (EMT) and, therefore, could contribute to renal fibrosis. In cultured tubular epithelial cells Gremlin binding to VEGFR2 is linked to proinflammatory responses. Now, we have found out that in these cells VEGFR2 is also involved in the profibrotic actions of Gremlin. VEGFR2 blockade by a pharmacological kinase inhibitor or gene silencing diminished Gremlin-mediated gene upregulation of profibrotic factors and restored changes in EMT-related genes. Moreover, VEGFR2 inhibition blocked EMT phenotypic changes and dampened the rate of wound healing in response to Gremlin. The role of VEGFR2 in experimental fibrosis was evaluated in experimental unilateral ureteral obstruction. VEFGR2 inhibition diminished the upregulation of profibrotic genes and EMT changes, as well as the accumulation of extracellular matrix proteins, such as fibronectin and collagens in the obstructed kidneys. Notch pathway activation participates in renal damage progression by regulating cell growth/proliferation, regeneration and inflammation. In cultured tubular epithelial cells, Notch inhibition markedly downregulated Gremlin-induced EMT changes and wound healing speed. These results show that Gremlin regulates the EMT process via VEGFR2 and Notch pathway activation, suggesting that the Gremlin/VEGFR2 axis could be a potential therapeutic target for CKD.
Collapse
Affiliation(s)
- Laura Marquez-Exposito
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain.,Red de Investigación Renal, Madrid, Spain
| | - Carolina Lavoz
- Division of Nephrology, School of Medicine, Universidad Austral, Valdivia, Chile
| | - Raul R Rodrigues-Diez
- Red de Investigación Renal, Madrid, Spain.,Laboratory of Nephrology, Fundación para la Investigación Biomédica del Hospital Universitario la Paz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Sandra Rayego-Mateos
- Red de Investigación Renal, Madrid, Spain.,Vascular and Renal Translational Research Group, Institut de Recerca Biomédica de Lleida, Lleida, Spain
| | - Macarena Orejudo
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain.,Red de Investigación Renal, Madrid, Spain
| | - Elena Cantero-Navarro
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain.,Red de Investigación Renal, Madrid, Spain
| | - Alberto Ortiz
- Red de Investigación Renal, Madrid, Spain.,Division of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Jesús Egido
- Division of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, Madrid, Spain
| | - Rafael Selgas
- Red de Investigación Renal, Madrid, Spain.,Laboratory of Nephrology, Fundación para la Investigación Biomédica del Hospital Universitario la Paz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Sergio Mezzano
- Division of Nephrology, School of Medicine, Universidad Austral, Valdivia, Chile
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain.,Red de Investigación Renal, Madrid, Spain
| |
Collapse
|
11
|
Gremlin activates the Notch pathway linked to renal inflammation. Clin Sci (Lond) 2018; 132:1097-1115. [PMID: 29720422 DOI: 10.1042/cs20171553] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 04/27/2018] [Accepted: 05/02/2018] [Indexed: 02/07/2023]
Abstract
Preclinical studies suggest that Gremlin participates in renal damage and could be a potential therapeutic target for human chronic kidney diseases. Inflammation is a common characteristic of progressive renal disease, and therefore novel anti-inflammatory therapeutic targets should be investigated. The Notch signaling pathway is involved in kidney development and is activated in human chronic kidney disease, but whether Gremlin regulates the Notch pathway has not been investigated. In cultured tubular cells, Gremlin up-regulated gene expression of several Notch pathway components, increased the production of the canonical ligand Jagged-1, and caused the nuclear translocation of active Notch-1 (N1ICD). In vivo administration of Gremlin into murine kidneys elicited Jagged-1 production, increased N1ICD nuclear levels, and up-regulated the gene expression of the Notch effectors hes-1 and hey-1 All these data clearly demonstrate that Gremlin activates the Notch pathway in the kidney. Notch inhibition using the γ-secretase inhibitor DAPT impaired renal inflammatory cell infiltration and proinflammatory cytokines overexpression in Gremlin-injected mice and in experimental models of renal injury. Moreover, Notch inhibition blocked Gremlin-induced activation of the canonical and noncanonical nuclear factor-κB (NF-κB) pathway, identifying an important mechanism involved in the anti-inflammatory actions of Notch inhibition. In conclusion, Gremlin activates the Notch pathway in the kidney and this is linked to NF-κB-mediated inflammation, supporting the hypothesis that Notch inhibition could be a potential anti-inflammatory strategy for renal diseases.
Collapse
|
12
|
Shaik S, Kennis B, Maegawa S, Schadler K, Yanwen Y, Callegari K, Lulla RR, Goldman S, Nazarian J, Rajaram V, Fangusaro J, Gopalakrishnan V. REST upregulates gremlin to modulate diffuse intrinsic pontine glioma vasculature. Oncotarget 2018; 9:5233-5250. [PMID: 29435175 PMCID: PMC5797046 DOI: 10.18632/oncotarget.23750] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 12/16/2017] [Indexed: 12/30/2022] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a highly aggressive glial tumor that occurs in children. The extremely poor median and 5-year survival in children afflicted with DIPG highlights the need for novel biology-driven therapeutics. Here, we have implicated the chromatin remodeler and regulator of brain development called RE1 Silencing Transcription Factor (REST), in DIPG pathology. We show that REST protein is aberrantly elevated in at least 21% of DIPG tumors compared to normal controls. Its knockdown in DIPG cell lines diminished cell growth and decreased their tumorigenicity in mouse intracranial models. DIPGs are vascularized tumors and interestingly, REST loss in DIPG cells also caused a substantial decline in tumor vasculature as measured by a decrease in CD31 and VEGFR2 staining. These observations were validated in vitro, where a significant decline in tube formation by human umbilical vein endothelial cells (HUVEC) was seen following REST-loss in DIPG cells. Mechanistically, REST controlled the secretion of a pro-angiogenic molecule and ligand for VEGFR2 called Gremlin-1 (GREM-1), and was associated with enhanced AKT activation. Importantly, the decline in tube formation caused by REST loss could be rescued by addition of recombinant GREM-1, which also caused AKT activation in HUVECs and human brain microvascular endothelial cells (HBMECs). In summary, our study is the first to demonstrate autocrine and paracrine functions for REST in DIPG development. It also provides the foundation for future investigations on anti-angiogenic therapies targeting GREM-1 in combination with drugs that target REST-associated chromatin remodeling activities.
Collapse
Affiliation(s)
- Shavali Shaik
- Department of Pediatrics, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Bridget Kennis
- Department of Pediatrics, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Shinji Maegawa
- Department of Pediatrics, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Keri Schadler
- Department of Pediatrics, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Yang Yanwen
- Department of Pediatrics, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Keri Callegari
- Department of Pediatrics, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Rishi R. Lulla
- Department of Pediatrics, Northwestern Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Stewart Goldman
- Department of Pediatrics, Northwestern Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Javad Nazarian
- Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Veena Rajaram
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jason Fangusaro
- Department of Pediatrics, Northwestern Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Vidya Gopalakrishnan
- Department of Pediatrics, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
- Department of Molecular and Cellular Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
- Center for Cancer Epigenetics, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
- Brain Tumor Center, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
13
|
Regulation of Vegf signaling by natural and synthetic ligands. Blood 2016; 128:2359-2366. [PMID: 27557946 DOI: 10.1182/blood-2016-04-711192] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 08/08/2016] [Indexed: 01/01/2023] Open
Abstract
The mechanisms that allow cells to bypass anti-vascular endothelial growth factor A (VEGFA) therapy remain poorly understood. Here we use zebrafish to investigate this question and first show that vegfaa mutants display a severe vascular phenotype that can surprisingly be rescued to viability by vegfaa messenger RNA injections at the 1-cell stage. Using vegfaa mutants as an in vivo test tube, we found that zebrafish Vegfbb, Vegfd, and Pgfb can also rescue these animals to viability. Taking advantage of a new vegfr1 tyrosine kinase-deficient mutant, we determined that Pgfb rescues vegfaa mutants via Vegfr1. Altogether, these data reveal potential resistance routes against current anti-VEGFA therapies. In order to circumvent this resistance, we engineered and validated new dominant negative Vegfa molecules that by trapping Vegf family members can block vascular development. Thus, our results show that Vegfbb, Vegfd, and Pgfb can sustain vascular development in the absence of VegfA, and our newly engineered Vegf molecules expand the toolbox for basic research and antiangiogenic therapy.
Collapse
|
14
|
Eikrem O, Beisland C, Hjelle K, Flatberg A, Scherer A, Landolt L, Skogstrand T, Leh S, Beisvag V, Marti HP. Transcriptome Sequencing (RNAseq) Enables Utilization of Formalin-Fixed, Paraffin-Embedded Biopsies with Clear Cell Renal Cell Carcinoma for Exploration of Disease Biology and Biomarker Development. PLoS One 2016; 11:e0149743. [PMID: 26901863 PMCID: PMC4764764 DOI: 10.1371/journal.pone.0149743] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 02/04/2016] [Indexed: 12/28/2022] Open
Abstract
Formalin-fixed, paraffin-embedded (FFPE) tissues are an underused resource for molecular analyses. This proof of concept study aimed to compare RNAseq results from FFPE biopsies with the corresponding RNAlater® (Qiagen, Germany) stored samples from clear cell renal cell carcinoma (ccRCC) patients to investigate feasibility of RNAseq in archival tissue. From each of 16 patients undergoing partial or full nephrectomy, four core biopsies, such as two specimens with ccRCC and two specimens of adjacent normal tissue, were obtained with a 16g needle. One normal and one ccRCC tissue specimen per patient was stored either in FFPE or RNAlater®. RNA sequencing libraries were generated applying the new Illumina TruSeq® Access library preparation protocol. Comparative analysis was done using voom/Limma R-package. The analysis of the FFPE and RNAlater® datasets yielded similar numbers of detected genes, differentially expressed transcripts and affected pathways. The FFPE and RNAlater datasets shared 80% (n = 1106) differentially expressed genes. The average expression and the log2 fold changes of these transcripts correlated with R2 = 0.97, and R2 = 0.96, respectively. Among transcripts with the highest fold changes in both datasets were carbonic anhydrase 9 (CA9), neuronal pentraxin-2 (NPTX2) and uromodulin (UMOD) that were confirmed by immunohistochemistry. IPA revealed the presence of gene signatures of cancer and nephrotoxicity, renal damage and immune response. To simulate the feasibility of clinical biomarker studies with FFPE samples, a classifier model was developed for the FFPE dataset: expression data for CA9 alone had an accuracy, specificity and sensitivity of 94%, respectively, and achieved similar performance in the RNAlater dataset. Transforming growth factor-ß1 (TGFB1)-regulated genes, epithelial to mesenchymal transition (EMT) and NOTCH signaling cascade may support novel therapeutic strategies. In conclusion, in this proof of concept study, RNAseq data obtained from FFPE kidney biopsies are comparable to data obtained from fresh stored material, thereby expanding the utility of archival tissue specimens.
Collapse
Affiliation(s)
- Oystein Eikrem
- Department of Clinical Medicine, Nephrology, University of Bergen, Bergen, Norway
| | - Christian Beisland
- Department of Clinical Medicine, Urology, University of Bergen, Bergen, Norway
| | - Karin Hjelle
- Department of Clinical Medicine, Urology, University of Bergen, Bergen, Norway
| | - Arnar Flatberg
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Lea Landolt
- Department of Clinical Medicine, Nephrology, University of Bergen, Bergen, Norway
| | - Trude Skogstrand
- Department of Clinical Medicine, Nephrology, University of Bergen, Bergen, Norway
| | - Sabine Leh
- Department of Clinical Medicine, Nephrology, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Vidar Beisvag
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Hans-Peter Marti
- Department of Clinical Medicine, Nephrology, University of Bergen, Bergen, Norway
- Department of Medicine, Nephrology, Haukeland University Hospital, Bergen, Norway
- * E-mail:
| |
Collapse
|
15
|
Yin Y, Yang Y, Yang L, Yang Y, Li C, Liu X, Qu Y. Overexpression of Gremlin promotes non-small cell lung cancer progression. Tumour Biol 2015; 37:2597-602. [PMID: 26392110 DOI: 10.1007/s13277-015-4093-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 09/13/2015] [Indexed: 10/23/2022] Open
Abstract
Lung cancer is the major cause of cancer-related death worldwide, and 80 % of them are non-small cell lung cancer (NSCLC) cases. Gremlin, a bone morphogenetic protein (BMP) antagonist, is overexpressed in various cancerous tissues; however, little is known about the roles of Gremlin in lung carcinogenesis, and it remains unclear whether Gremlin expression may associate with EGFR-TKI resistance. In this study, expression of Gremlin mRNA and protein in matched tumor and normal lung specimens are quantified by quantitative real-time PCR and western blot. The functional role of Gremlin in NSCLC cells was evaluated by interference RNA (siRNA). The effects of Silenced Gremlin on the resistant PC-9/GR cell line were investigated by proliferation and apoptosis analysis compared with control PC-9 cells. Our results found that Gremlin expression levels were higher in NSCLC tissues, and Gremlin was more highly expressed in PC-9/GR cells compared to PC-9 cells. Knocking down of Gremlin in PC-9/GR cells decreased cell proliferation and increased the expression of BMP7 protein. In addition, Gremlin silencing significantly potentiated apoptosis induced by gefitinib in PC-9/GR with Gremlin knockdown compared to PC-9 transfected with control shRNA, suggesting Gremlin contributes to gefitinib resistance in NSCLC. Gremlin might be explored as a candidate of therapeutic target for modulating EGFR-TKI sensitivity in NSCLC.
Collapse
Affiliation(s)
- Yunhong Yin
- Department of Respiratory Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Yie Yang
- Clinical Laboratory, Qianfoshan Hospital of Shandong Province, Jinan, 250012, China
| | - Liyun Yang
- Yinan Branch of Qilu Hospital of Shandong University, Yinan, 276300, China
| | - Yan Yang
- Department of Respiratory Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Chunyu Li
- Department of Respiratory Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Xiao Liu
- Department of Respiratory Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Yiqing Qu
- Department of Respiratory Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China.
| |
Collapse
|