Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters

Publication year range
1.
J Neuroinflammation ; 20(1): 116, 2023 May 16.
Article in English | MEDLINE | ID: mdl-37194065

ABSTRACT

Discoidin Domain Receptor (DDR)-1 is activated by collagen. Nilotinib is a tyrosine kinase inhibitor that is FDA-approved for leukemia and potently inhibits DDR-1. Individuals diagnosed with mild-moderate Alzheimer's disease (AD) treated with nilotinib (versus placebo) for 12 months showed reduction of amyloid plaque and cerebrospinal fluid (CSF) amyloid, and attenuation of hippocampal volume loss. However, the mechanisms are unclear. Here, we explored unbiased next generation whole genome miRNA sequencing from AD patients CSF and miRNAs were matched with their corresponding mRNAs using gene ontology. Changes in CSF miRNAs were confirmed via measurement of CSF DDR1 activity and plasma levels of AD biomarkers. Approximately 1050 miRNAs are detected in the CSF but only 17 miRNAs are specifically altered between baseline and 12-month treatment with nilotinib versus placebo. Treatment with nilotinib significantly reduces collagen and DDR1 gene expression (upregulated in AD brain), in association with inhibition of CSF DDR1. Pro-inflammatory cytokines, including interleukins and chemokines are reduced along with caspase-3 gene expression. Specific genes that indicate vascular fibrosis, e.g., collagen, Transforming Growth Factors (TGFs) and Tissue Inhibitors of Metalloproteases (TIMPs) are altered by DDR1 inhibition with nilotinib. Specific changes in vesicular transport, including the neurotransmitters dopamine and acetylcholine, and autophagy genes, including ATGs, indicate facilitation of autophagic flux and cellular trafficking. Inhibition of DDR1 with nilotinib may be a safe and effective adjunct treatment strategy involving an oral drug that enters the CNS and adequately engages its target. DDR1 inhibition with nilotinib exhibits multi-modal effects not only on amyloid and tau clearance but also on anti-inflammatory markers that may reduce cerebrovascular fibrosis.


Subject(s)
Alzheimer Disease , MicroRNAs , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Discoidin Domain Receptors , Pyrimidines/pharmacology , Collagen/therapeutic use , Fibrosis , Inflammation/drug therapy
2.
J Comput Aided Mol Des ; 37(11): 507-517, 2023 11.
Article in English | MEDLINE | ID: mdl-37550462

ABSTRACT

Generative approaches to molecular design are an area of intense study in recent years as a method to generate new pharmaceuticals with desired properties. Often though, these types of efforts are constrained by limited experimental activity data, resulting in either models that generate molecules with poor performance or models that are overfit and produce close analogs of known molecules. In this paper, we reduce this data dependency for the generation of new chemotypes by incorporating docking scores of known and de novo molecules to expand the applicability domain of the reward function and diversify the compounds generated during reinforcement learning. Our approach employs a deep generative model initially trained using a combination of limited known drug activity and an approximate docking score provided by a second machine learned Bayes regression model, with final evaluation of high scoring compounds by a full docking simulation. This strategy results in molecules with docking scores improved by 10-20% compared to molecules of similar size, while being 130 × faster than a docking only approach on a typical GPU workstation. We also show that the increased docking scores correlate with (1) docking poses with interactions similar to known inhibitors and (2) result in higher MM-GBSA binding energies comparable to the energies of known DDR1 inhibitors, demonstrating that the Bayesian model contains sufficient information for the network to learn to efficiently interact with the binding pocket during reinforcement learning. This outcome shows that the combination of the learned latent molecular representation along with the feature-based docking regression is sufficient for reinforcement learning to infer the relationship between the molecules and the receptor binding site, which suggest that our method can be a powerful tool for the discovery of new chemotypes with potential therapeutic applications.


Subject(s)
Deep Learning , Drug Discovery , Bayes Theorem , Computer Simulation , Machine Learning , Drug Design
3.
J Pathol ; 258(1): 26-37, 2022 09.
Article in English | MEDLINE | ID: mdl-35607980

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 α2ß1 integrin/DDR1 co-receptor signaling as the dominant regulatory mechanism. This may explain earlier studies where deletion of either DDR1 or α2ß1 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.


Subject(s)
Nephritis, Hereditary , Podocytes , Animals , Basement Membrane/pathology , Collagen Type III , Collagen Type IV/genetics , Discoidin Domain Receptor 1/genetics , Glomerular Basement Membrane/pathology , Humans , Integrin alpha2beta1 , Mice , Mice, Knockout , Nephritis, Hereditary/genetics , Nephritis, Hereditary/pathology , Podocytes/pathology , Pseudopodia/pathology , RNA
4.
Mol Divers ; 27(5): 2297-2314, 2023 Oct.
Article in English | MEDLINE | ID: mdl-36322341

ABSTRACT

Discoidin domain receptor 1 (DDR1) (EC Number 2.7.10.1) has recently been considered as a promising therapeutic target for idiopathic pulmonary fibrosis (IPF). However, none of the currently discovered DDR1 inhibitors have been included in clinical studies due to low target specificity or druggability limitations, necessitating various approaches to develop novel DDR1 inhibitors. In this study, to assure target specificity, a docking assessment of the DDR1 crystal structures was undertaken to find the well-differentiated crystal structure, and 4CKR was identified among many crystal structures. Then, using the best pharmacophore model and molecular docking, virtual screening of the ChEMBL database was done, and five potential molecules were identified as promising inhibitors of DDR1. Subsequently, all hit compound complex systems were validated using molecular dynamics simulations and MM/PBSA methods to assess the stability of the system after ligand binding to DDR1. Based on molecular dynamics simulations and hydrogen-bonding occupancy analysis, the DDR1-Cpd2, DDR1-Cpd17, and DDR1-Cpd18 complex systems exhibited superior stability compared to the DDR1-Cpd1 and DDR-Cpd33 complex systems. Meanwhile, when targeting DDR1, the descending order of the five hit molecules' binding free energies was Cpd17 (- 145.820 kJ/mol) > Cpd2 (- 131.818 kJ/mol) > Cpd18 (- 130.692 kJ/mol) > Cpd33 (- 129.175 kJ/mol) > Cpd1 (- 126.103 kJ/mol). Among them, Cpd2, Cpd17, and Cpd18 showed improved binding characteristics, indicating that they may be potential DDR1 inhibitors. In this research, we developed a high-hit rate, effective screening method that serves as a theoretical guide for finding DDR1 inhibitors for the development of IPF therapeutics.


Subject(s)
Discoidin Domain Receptor 1 , Receptor Protein-Tyrosine Kinases , Receptor Protein-Tyrosine Kinases/chemistry , Discoidin Domain Receptors , Receptors, Mitogen/chemistry , Receptors, Mitogen/metabolism , Molecular Docking Simulation
5.
J Periodontal Res ; 57(3): 568-577, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35297053

ABSTRACT

BACKGROUND AND OBJECTIVE: As one of the widely expressed cell surface receptors binding to collagen, the most abundant component of the extracellular matrix (ECM), knowledge of the expression, functions, and mechanisms underlying the role of discoidin domain receptor 1 (DDR1) in human periodontal ligament cells (hPDLCs) is incomplete. This study determined the expression of DDR1 in hPDLCs and the effect of DDR1 upon migration and adhesion to hPDLCs, as well as the related regulatory mechanisms. MATERIALS AND METHODS: The expression of DDR1 and the DDR1 isoforms in hPDLCs from six donors were tested. The migratory ability (horizontal and vertical) and adhesive capacity of hPDLCs with or without specific knockdown of DDR1 were evaluated. After treatment with MEK-ERK1/2 inhibitors (PD98059 and U0126) with or without RNAi, the migratory and adhesive capacity of hPDLCs were re-tested. Western blotting was performed to verify p-MEK1/2 and p-ERK1/2, the key factors of the MEK-ERK1/2 signaling pathways. RESULTS: DDR1 was detected in hPDLCs in the mRNA and protein level; DDR1b was the dominant isoform. Knockdown of DDR1 almost halved the migratory capacity and significantly downregulated the adhesive capacity of hPDLCs. The use of MEK-ERK1/2 inhibitors caused declined migratory and adhesive capacity of hPDLCs as well. After DDR1 was knocked down, the expression of p-MEK and p-ERK protein declined significantly while total MEK and ERK showed no obvious change, which means the ratio of p-MEK/MEK and p-ERK/ERK was markedly reduced. CONCLUSIONS: DDR1 plays an important role in the migration and adhesion of hPDLCs and might be regulated via the MEK-ERK1/2 signaling pathway.


Subject(s)
Discoidin Domain Receptor 1 , Periodontal Ligament , Cell Adhesion , Cell Movement , Cells, Cultured , Discoidin Domain Receptor 1/metabolism , Humans , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism
6.
Am J Physiol Gastrointest Liver Physiol ; 320(6): G1002-G1013, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33851541

ABSTRACT

Tumor DDR1 acts as a key factor during the desmoplastic response surrounding hepatic colorectal metastasis. Hepatic sinusoidal cell-derived soluble factors stimulate tumor DDR1 activation. DDR1 modulates matrix remodeling to promote metastasis in the liver through the interaction with hepatic stromal cells, specifically liver sinusoidal endothelial cells and hepatic stellate cells.


Subject(s)
Carcinoma/genetics , Colonic Neoplasms/genetics , Discoidin Domain Receptor 1/genetics , Liver Neoplasms/genetics , Liver/pathology , Animals , Carcinoma/metabolism , Carcinoma/secondary , Cell Line, Tumor , Cell Proliferation , Colon/metabolism , Colon/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Discoidin Domain Receptor 1/metabolism , Hepatic Stellate Cells/pathology , Humans , Liver/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Male , Mice , Phosphorylation , Prognosis , Stromal Cells/metabolism , Stromal Cells/pathology
7.
FASEB J ; 34(4): 5767-5781, 2020 04.
Article in English | MEDLINE | ID: mdl-32128899

ABSTRACT

Chondrocytes in growth plates are responsible for longitudinal growth in long bones during endochondral ossification. Discoidin domain receptor 1 (Ddr1) is expressed in chondrocytes, but the molecular mechanisms by which DDR1 regulates chondrocyte behaviors during the endochondral ossification process remain undefined. To elucidate Ddr1-mediate chondrocyte functions, we generated chondrocyte-specific Ddr1 knockout (CKOΔDdr1) mice in this study. The CKOΔDdr1 mice showed delayed development of the secondary ossification center and increased growth plate length in the hind limbs. In the tibial growth plate in CKOΔDdr1 mice, chondrocyte proliferation was reduced in the proliferation zone, and remarkable downregulation of Ihh, MMP13, and Col-X expression in chondrocytes resulted in decreased terminal differentiation in the hypertrophic zone. Furthermore, apoptotic chondrocytes were reduced in the growth plates of CKOΔDdr1 mice. We concluded that chondrocytes with Ddr1 knockout exhibit decreased proliferation, terminal differentiation, and apoptosis in growth plates, which delays endochondral ossification and results in short stature. We also demonstrated that Ddr1 regulates the Ihh/Gli1/Gli2/Col-X pathway to regulate chondrocyte terminal differentiation. These results indicate that Ddr1 is required for chondrocytes to regulate endochondral ossification in skeletal development.


Subject(s)
Bone and Bones/cytology , Cell Differentiation , Chondrocytes/cytology , Chondrogenesis , Discoidin Domain Receptor 1/physiology , Osteogenesis , Animals , Chondrocytes/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
8.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 43(4): 634-641, 2021 Aug.
Article in Zh | MEDLINE | ID: mdl-34494537

ABSTRACT

Discoidin domain receptor 1(DDR1)is a critical member of the receptor tyrosine kinase family.It may be related to tumor invasion and metastasis,and the abnormal activation of DDR1 can lead to the occurrence and development of malignant tumors,inflammation,and fibrosis.DDR1 are involved in cell adhesion,migration,proliferation,secretion of cytokines,and remodeling of extracellular matrix,thus playing a critical role in various pathophysiological processes of the human body.In this review,we demonstrate the research progress of DDR1 in breast cancer and other malignant tumors,in order to provide a new theoretical basis for the prevention and treatment of breast cancer and other tumors.


Subject(s)
Breast Neoplasms , Discoidin Domain Receptor 1 , Breast Neoplasms/genetics , Cell Adhesion , Female , Fibrosis , Humans , Receptor Protein-Tyrosine Kinases/genetics
9.
Int J Mol Sci ; 21(19)2020 Sep 29.
Article in English | MEDLINE | ID: mdl-33003599

ABSTRACT

Discoidin domain receptor 1 (Drd1) is a collagen-binding membrane protein, but its role in osteoblasts during osteogenesis remains undefined. We generated inducible osteoblast-specific Ddr1 knockout (OKOΔDdr1) mice; their stature at birth, body weight and body length were significantly decreased compared with those of control Ddr1f/f-4OHT mice. We hypothesize that Ddr1 regulates osteogenesis of osteoblasts. Micro-CT showed that compared to 4-week-old Ddr1f/f-4OHT mice, OKOΔDdr1 mice presented significant decreases in cancellous bone volume and trabecular number and significant increases in trabecular separation. The cortical bone volume was decreased in OKOΔDdr1 mice, resulting in decreased mechanical properties of femurs compared with those of Ddr1f/f-4OHT mice. In femurs of 4-week-old OKOΔDdr1 mice, H&E staining showed fewer osteocytes and decreased cortical bone thickness than Ddr1f/f-4OHT. Osteoblast differentiation markers, including BMP2, Runx2, alkaline phosphatase (ALP), Col-I and OC, were decreased compared with those of control mice. Ddr1 knockdown in osteoblasts resulted in decreased mineralization, ALP activity, phosphorylated p38 and protein levels of BMP2, Runx2, ALP, Col-I and OC during osteogenesis. Overexpression and knockdown of Ddr1 in osteoblasts demonstrated that DDR1 mediates the expression and activity of Runx2 and the downstream osteogenesis markers during osteogenesis through regulation of p38 phosphorylation.


Subject(s)
Core Binding Factor Alpha 1 Subunit/genetics , Osteogenesis/genetics , Receptors, Dopamine D1/genetics , p38 Mitogen-Activated Protein Kinases/genetics , Alkaline Phosphatase/genetics , Animals , Bone Morphogenetic Protein 2/genetics , Collagen/genetics , Femur/growth & development , Femur/metabolism , Gene Expression Regulation, Developmental/genetics , Mice , Mice, Knockout , Osteoblasts/metabolism , Phosphorylation/genetics
10.
J Biol Chem ; 293(8): 2841-2849, 2018 02 23.
Article in English | MEDLINE | ID: mdl-29298894

ABSTRACT

Discoidin domain receptor 1 (DDR1) is a collagen receptor that mediates cell communication with the extracellular matrix (ECM). Aberrant expression and activity of DDR1 in tumor cells are known to promote tumor growth. Although elevated DDR1 levels in the stroma of breast tumors are associated with poor patient outcome, a causal role for tumor-extrinsic DDR1 in cancer promotion remains unclear. Here we report that murine mammary tumor cells transplanted to syngeneic recipient mice in which Ddr1 has been knocked out (KO) grow less robustly than in WT mice. We also found that the tumor-associated stroma in Ddr1-KO mice exhibits reduced collagen deposition compared with the WT controls, supporting a role for stromal DDR1 in ECM remodeling of the tumor microenvironment. Furthermore, the stromal-vascular fraction (SVF) of Ddr1 knockout adipose tissue, which contains committed adipose stem/progenitor cells and preadipocytes, was impaired in its ability to stimulate tumor cell migration and invasion. Cytokine array-based screening identified interleukin 6 (IL-6) as a cytokine secreted by the SVF in a DDR1-dependent manner. SVF-produced IL-6 is important for SVF-stimulated tumor cell invasion in vitro, and, using antibody-based neutralization, we show that tumor promotion by IL-6 in vivo requires DDR1. In conclusion, our work demonstrates a previously unrecognized function of DDR1 in promoting tumor growth.


Subject(s)
Adipose Tissue/metabolism , Breast Neoplasms/metabolism , Discoidin Domain Receptor 1/metabolism , Interleukin-6/metabolism , Stromal Cells/metabolism , Adipose Tissue/drug effects , Adipose Tissue/immunology , Adipose Tissue/pathology , Animals , Antibodies, Neutralizing/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Collagen/metabolism , Discoidin Domain Receptor 1/genetics , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Invasiveness/immunology , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Stromal Cells/drug effects , Stromal Cells/immunology , Stromal Cells/pathology , Transplantation, Isogeneic , Tumor Burden/drug effects , Tumor Cells, Cultured , Tumor Microenvironment/drug effects
11.
J Cell Physiol ; 234(11): 19539-19552, 2019 11.
Article in English | MEDLINE | ID: mdl-30963567

ABSTRACT

Didscoidin domain receptor 1 (DDR1) is involved in the progression of prostate cancer metastasis through stimulation of epithelial-mesenchymal transition (EMT). So DDR1 inhibition can be a helpful target for cancer metastasis prevention. So, we studied the effects of DDR1 inhibition on EMT as well as induction of cell-cycle arrest and apoptosis in prostate cancer cell lines. DDR1 expression was evaluated using reverse-transcription polymerase chain reaction and western blot analysis. The EMT-associated protein expression was determined using the western blot analysis and immunocytochemistry following treatment with various concentrations of DDR1 inhibitor. The activation of DDR1 and also downstream-signaling molecules Pyk2 and MKK7 were determined using western blot analysis. Cell survival and proliferation after DDR1 inhibition were evaluated using 3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyltetrazolium bromide, bromodeoxyuridine, and colony formation assays. Flow cytometry analysis was used to determine the effects of DDR1 inhibition on cell-cycle arrest and apoptosis using annexin V/propidium iodide-based flow cytometry. Results showed that the protein expression of N-cadherin and vimentin were decreased whereas protein expression of E-cadherin was increased after DDR1 inhibition. Results of our western blot analysis indicated that DDR1 inhibitor effectively downregulated P-DDR1, P-Pyk2, and P-MKK7 levels. This result also showed that DDR1 inhibition decreased cell survival and proliferation, induced G1 cell-cycle arrest, induced apoptosis by an increase in the Bax/Bcl-2 ratio and depletion of the mitochondrial membrane potential, and also by reactive oxygen species creation in prostate cancer cells. These data show that DDR1 inhibition can result in the EMT prevention via inhibition of Pyk2 and MKK7 signaling pathway and induces cell-cycle arrest and apoptosis in prostate cancer cell lines. Thus, this study identifies DDR1 as an important target for modulating EMT and induction of apoptosis in prostate cancer cells.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/genetics , Discoidin Domain Receptor 1/genetics , Prostatic Neoplasms/genetics , Antineoplastic Agents , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Discoidin Domain Receptor 1/antagonists & inhibitors , Epithelial-Mesenchymal Transition/genetics , Focal Adhesion Kinase 2/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , MAP Kinase Kinase 7/genetics , Male , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction/genetics , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , bcl-2-Associated X Protein/genetics
12.
Arterioscler Thromb Vasc Biol ; 38(8): 1878-1889, 2018 08.
Article in English | MEDLINE | ID: mdl-29930002

ABSTRACT

Objective- Vascular calcification is a common and severe complication in patients with atherosclerosis which is exacerbated by type 2 diabetes mellitus. Our laboratory recently reported that the collagen receptor discoidin domain receptor 1 (DDR1) mediates vascular calcification in atherosclerosis; however, the underlying mechanisms are unknown. During calcification, vascular smooth muscle cells transdifferentiate into osteoblast-like cells, in a process driven by the transcription factor RUNX2 (runt-related transcription factor 2). DDR1 signals via the phosphoinositide 3-kinase/Akt pathway, which is also central to insulin signaling, and upstream of RUNX2, and this led us to investigate whether DDR1 promotes vascular calcification in diabetes mellitus via this pathway. Approach and Results- Ddr1+/+ ; Ldlr-/- (single knock-out) and Ddr1-/- ; Ldlr-/- (double knock-out) mice were placed on high-fat diet for 12 weeks to induce atherosclerosis and type 2 diabetes mellitus. Von Kossa staining revealed reduced vascular calcification in the aortic arch of double knock-out compared with single knock-out mice. Immunofluorescent staining for RUNX2 was present in calcified plaques of single knock-out but not double knock-out mice. Primary vascular smooth muscle cells obtained from Ddr1+/+ and Ddr1-/- mice were cultured in calcifying media. DDR1 deletion resulted in reduced calcification, a 74% reduction in p-Akt levels, and an 88% reduction in RUNX2 activity. Subcellular fractionation revealed a 77% reduction in nuclear RUNX2 levels in Ddr1-/- vascular smooth muscle cells. DDR1 associated with phosphoinositide 3-kinase, and treatment with the inhibitor wortmannin attenuated calcification. Finally, we show that DDR1 is important to maintain the microtubule cytoskeleton which is required for the nuclear localization of RUNX2. Conclusions- These novel findings demonstrate that DDR1 promotes RUNX2 activity and atherosclerotic vascular calcification in diabetes mellitus via phosphoinositide 3-kinase/Akt signaling.


Subject(s)
Atherosclerosis/enzymology , Core Binding Factor Alpha 1 Subunit/metabolism , Diabetes Mellitus, Type 2/enzymology , Diabetic Angiopathies/enzymology , Discoidin Domain Receptor 1/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Vascular Calcification/enzymology , Active Transport, Cell Nucleus , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Cells, Cultured , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Diabetic Angiopathies/genetics , Diabetic Angiopathies/pathology , Diet, High-Fat , Discoidin Domain Receptor 1/deficiency , Discoidin Domain Receptor 1/genetics , Disease Models, Animal , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Phosphorylation , Receptors, LDL/deficiency , Receptors, LDL/genetics , Signal Transduction , Vascular Calcification/genetics , Vascular Calcification/pathology
13.
BMC Cancer ; 17(1): 87, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28143619

ABSTRACT

BACKGROUND: Discoidin domain receptor 1 (DDR1), a receptor tyrosine kinase that utilizes collagen as a ligand, is a key molecule in the progression of solid tumors as it regulates the interaction of cancer cells with the tumor stroma. However, the clinical relevance of DDR1 expression in gastric carcinoma is yet to be investigated. Here, we assessed the role of DDR1 in mediating the aggressive phenotype of gastric carcinoma and its potential as a therapeutic target. METHODS: We conducted DDR1 immunohistochemistry using a tissue microarray of 202 gastric carcinoma specimens. We examined the effect of collagen-induced activation of DDR1 on cell signaling, tumorigenesis, and cell migration in gastric cancer cell lines, and tumor growth in a xenograft animal model of gastric cancer. RESULTS: Our results showed that 50.5% of gastric cancer tissues are positive for DDR1 expression, and positive DDR1 expression was significantly correlated with a poor prognosis (P = 0.015). In a subgroup analysis, DDR1 expression was prognostically meaningful only in patients receiving adjuvant treatment (P = 0.013). We also demonstrated that collagen was able to activate DDR1 and increase the clonogenicity and migration of gastric cancer cells. We observed that a DDR1 inhibitor, 7rh benzamide, suppressed tumor growth in gastric cancer xenografts. CONCLUSIONS: Our findings suggest a key role for DDR1 signaling in mediating the aggressive phenotype of gastric carcinoma. Importantly, inhibition of DDR1 is an attractive strategy for gastric carcinoma therapy.


Subject(s)
Carcinoma/genetics , Carcinoma/pathology , Discoidin Domain Receptor 1/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Cell Movement/genetics , Collagen/metabolism , Humans , Immunohistochemistry/methods , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Phenotype , Prognosis , Signal Transduction/genetics
14.
Microsc Microanal ; 22(3): 599-611, 2016 06.
Article in English | MEDLINE | ID: mdl-27329311

ABSTRACT

The quantity and quality of collagen fibrils in the extracellular matrix (ECM) have a pivotal role in dictating biological processes. Several collagen-binding proteins (CBPs) are known to modulate collagen deposition and fibril diameter. However, limited studies exist on alterations in the fibril ultrastructure by CBPs. In this study, we elucidate how the collagen receptor, discoidin domain receptor 1 (DDR1) regulates the collagen content and ultrastructure in the adventitia of DDR1 knock-out (KO) mice. DDR1 KO mice exhibit increased collagen deposition as observed using Masson's trichrome. Collagen ultrastructure was evaluated in situ using transmission electron microscopy, scanning electron microscopy, and atomic force microscopy. Although the mean fibril diameter was not significantly different, DDR1 KO mice had a higher percentage of fibrils with larger diameter compared with their wild-type littermates. No significant differences were observed in the length of D-periods. In addition, collagen fibrils from DDR1 KO mice exhibited a small, but statistically significant, increase in the depth of the fibril D-periods. Consistent with these observations, a reduction in the depth of D-periods was observed in collagen fibrils reconstituted with recombinant DDR1-Fc. Our results elucidate how DDR1 modulates collagen fibril ultrastructure in vivo, which may have important consequences in the functional role(s) of the underlying ECM.


Subject(s)
Collagen/ultrastructure , Discoidin Domain Receptor 1/genetics , Extracellular Matrix/genetics , Animals , Discoidin Domain Receptor 1/metabolism , Mice , Mice, Knockout , Protein Binding
15.
Am J Physiol Cell Physiol ; 308(9): C685-96, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25673773

ABSTRACT

Effectively directing the chondrogenesis of adipose-derived stem cells (ADSCs) to engineer articular cartilage represents an important challenge in ADSC-based articular cartilage tissue engineering. The discoidin domain receptor 1 (DDR1) has been shown to affect cartilage homeostasis; however, little is known about the roles of DDR1 in ADSC chondrogenesis. In this study, we used the three-dimensional culture pellet culture model system with chondrogenic induction to investigate the roles of DDR1 in the chondrogenic differentiation of human ADSCs (hADSCs). Real-time polymerase chain reaction and Western blot were used to detect the expression of DDRs and chondrogenic genes. Sulfated glycosaminoglycan (sGAG) was detected by Alcian blue and dimethylmethylene blue (DMMB) assays. Terminal deoxy-nucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining was used to assess cell death. During the chondrogenesis of hADSCs, the expression of DDR1 but not DDR2 was significantly elevated. The depletion of DDR1 expression in hADSCs using short hairpin RNA increased the expression of chondrogenic genes (SOX-9, collagen type II, and aggrecan) and cartilaginous matrix deposition (collagen type II and sGAG) and only slightly increased cell death (2-8%). DDR1 overexpression in hADSCs decreased the expression of chondrogenic genes (SOX-9, collagen type II, and aggrecan) and sGAG and enhanced hADSC survival. Moreover, DDR1-depleted hADSCs showed decreased expression of the terminal differentiation genes runt-related transcription factor 2 (Runx2) and matrix metalloproteinase 13 (MMP-13). These results suggest that DDR1 suppression may enhance ADSC chondrogenesis by enhancing the expression of chondrogenic genes and cartilaginous matrix deposition. We proposed that the suppression of DDR1 in ADSCs may be a candidate strategy of genetic modification to optimize ADSC-based articular cartilage tissue engineering.


Subject(s)
Chondrocytes/metabolism , Chondrogenesis , Receptor Protein-Tyrosine Kinases/metabolism , Stem Cells/metabolism , Subcutaneous Fat/metabolism , Aggrecans/genetics , Aggrecans/metabolism , Cell Differentiation , Cell Survival , Cells, Cultured , Collagen Type II/genetics , Collagen Type II/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Discoidin Domain Receptor 1 , Gene Expression Regulation , Glycosaminoglycans/metabolism , Humans , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Phenotype , RNA Interference , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/genetics , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Signal Transduction , Subcutaneous Fat/cytology , Time Factors , Transfection
16.
Int J Cancer ; 136(6): E508-20, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25155634

ABSTRACT

The epithelial-to-mesenchymal transition (EMT) process allows carcinoma cells to dissociate from the primary tumor thereby facilitating tumor cell invasion and metastasis. Ras-dependent hyperactive signaling is commonly associated with tumorigenesis, invasion, EMT, and metastasis. However, the downstream effectors by which Ras regulates EMT remain ill defined. In this study, we show that the H-Ras pathway leads to mesenchymal-like phenotypic changes in human breast epithelial cells by controlling the ZEB1/microRNA-200c axis. Moreover, H-Ras suppresses the expression of the discoidin domain receptor 1 (DDR1), a collagen receptor tyrosine kinase, via ZEB1, thus identifying ZEB1 as a novel transcriptional repressor of DDR1. Mutation studies on the putative promoter of the DDR1 gene revealed that bipartite Z- and E-box elements play a key role in transcriptional repression of DDR1 in Hs578T and MDA-MB-231 breast carcinoma cell lines by ZEB1. Furthermore, we found an inverse correlation between ZEB1 and DDR1 expression in various cancer cell lines and in human breast carcinoma tissues. Consistently, overexpression of DDR1 reduced the invasive phenotype of mesenchymal-like triple-negative breast cancer cells in 3D cultures and in vivo. Thus, ZEB1's role in maintenance of EMT in breast carcinoma cells is mediated in part by its ability to suppress DDR1 expression and consequently contribute to the activation of the invasive phenotype. Taken together, our results unveil a novel H-Ras/ZEB1/DDR1 network that contributes to breast cancer progression in triple-negative breast cancers.


Subject(s)
Breast/pathology , Epithelial-Mesenchymal Transition , Genes, ras/physiology , Homeodomain Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Mitogen/physiology , Transcription Factors/physiology , Cell Line, Tumor , Cytoskeleton/physiology , Discoidin Domain Receptors , Epithelial Cells/pathology , Female , Humans , MicroRNAs/physiology , Morphogenesis , Zinc Finger E-box-Binding Homeobox 1
17.
Nephrol Dial Transplant ; 30(12): 1965-71, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25829327

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.


Subject(s)
Cell Adhesion Molecules/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Renal Insufficiency, Chronic/metabolism , Discoidin Domain Receptor 1 , Humans
18.
J Biol Chem ; 288(23): 16738-16746, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23530036

ABSTRACT

Growing evidence demonstrates that extracellular matrices regulate many aspects of megakaryocyte (MK) development; however, among the different extracellular matrix receptors, integrin α2ß1 and glycoprotein VI are the only collagen receptors studied in platelets and MKs. In this study, we demonstrate the expression of the novel collagen receptor discoidin domain receptor 1 (DDR1) by human MKs at both mRNA and protein levels and provide evidence of DDR1 involvement in the regulation of MK motility on type I collagen through a mechanism based on the activity of SHP1 phosphatase and spleen tyrosine kinase (Syk). Specifically, we demonstrated that inhibition of DDR1 binding to type I collagen, preserving the engagement of the other collagen receptors, glycoprotein VI, α2ß1, and LAIR-1, determines a decrease in MK migration due to the reduction in SHP1 phosphatase activity and consequent increase in the phosphorylation level of its main substrate Syk. Consistently, inhibition of Syk activity restored MK migration on type I collagen. In conclusion, we report the expression and function of a novel collagen receptor on human MKs, and we point out that an increasing level of complexity is necessary to better understand MK-collagen interactions in the bone marrow environment.


Subject(s)
Cell Movement/physiology , Collagen Type I/metabolism , Megakaryocytes/enzymology , Receptor Protein-Tyrosine Kinases/metabolism , CD36 Antigens/genetics , CD36 Antigens/metabolism , Collagen Type I/genetics , Discoidin Domain Receptor 1 , Female , Humans , Integrin alpha2beta1/genetics , Integrin alpha2beta1/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Megakaryocytes/cytology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Syk Kinase
19.
Nephrol Dial Transplant ; 29 Suppl 4: iv124-30, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25165179

ABSTRACT

The hereditary type IV collagen disease Alport syndrome (AS) always leads to end-stage renal failure. Yesterday, for the past 90 years, this course was described as 'inevitable'. Today, RAAS blockade has changed the 'inevitable' course to a treatable disease. Tomorrow, researchers hope to erase the 'always' from 'always leads to renal failure' in the textbooks. This review elucidates therapeutic targets that evolve from research: (i) kidney embryogenesis and pathogenesis; (ii) phenotype-genotype correlation and the role of collagen receptors and podocytes; (iii) the malfunctioning Alport-GBM; (iv) tubulointerstitial fibrosis; (v) the role of proteinuria in pathogenesis and prognosis; and (vi) secondary events such as infections, hyperparathyroidism and hypercholesterolaemia. Therefore, moderate lifestyle, therapy of bacterial infections, Paricalcitol in adult patients with hyperparathyroidism and HMG-CoA-reductase inhibitors in adult patients with dyslipoproteinemia might contribute to a slower progression of AS and less cardiovascular events. In the future, upcoming treatments including stem cells, chaperon therapy, collagen receptor blockade and anti-microRNA therapy will expand our perspective in protecting the kidneys of Alport patients from further damage. This perspective on current and future therapies is naturally limited by our personal focus in research, but aims to motivate young scientists and clinicians to find a multimodal cure for AS.


Subject(s)
Angiotensin Receptor Antagonists/therapeutic use , Nephritis, Hereditary/therapy , Renin-Angiotensin System/drug effects , Adult , Collagen Type IV/genetics , Humans , Nephritis, Hereditary/genetics
20.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167447, 2024 10.
Article in English | MEDLINE | ID: mdl-39089636

ABSTRACT

Gefitinib is an epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI), which serves the critical pillar for the treatment of non-small cell lung cancer (NSCLC). However, the acquired resistance remains a challenge for its clinical application, for which, practical strategies to reverse gefitinib resistance in NSCLC are necessary. Ferroptosis, a programmed cell death driven by ferritin-dependent lipid peroxidation, involves in NSCLC progression and related chemoresistance. In our previous work, the self-synthesised EGFR inhibitor Yfq07 (N4, N6-disubstituted pyrimidine-4,6-diamine derivatives) displayed a considerable inhibitory effect on NSCLC both in vitro and in vivo. Herein, we observed that Yfq07 suppressed the proliferation of PC-9GR and HCC827GR cells, two gefitinib resistance NSCLC cell lines. Mechanically, Yfq07 inhibited the phosphorylation of the Discoidin Domain Receptor 1 (DDR1), a receptor tyrosine kinase (RTK) highly expressed in multiple cancers, accompanied by downregulated miR-3648 and upregulated SOCS2. Inhibition or knockdown of DDR1 suppressed the proliferation, migration, and invasion of gefitinib-resistant NSCLC cells, and on the other hand, also downregulated miR-3648 and promoted SOCS2 expression. More specifically, miR-3648 targeted the 3'UTR segment of SOCS2 mRNA and thus affecting the P-ERK signalling pathway to regulate the malignant behaviors of gefitinib-resistant NSCLC cells. Furthermore, Yfq07 also indirectly induced the ferroptosis of gefitinib-resistant NSCLC cells via SOCS2 triggered inhibition of xCT-GPX4 pathway. In conclusion, our study indicates that DDR1 inhibitor Yfq07 promotes ferroptosis and reverses gefitinib-resistance of NSCLC through DDR1-miR-3648-SOCS2 signalling pathway, which provides insights for targeted therapy of gefitinib-resistant NSCLC and drug developments targeting ferroptosis.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Discoidin Domain Receptor 1 , Drug Resistance, Neoplasm , Ferroptosis , Gefitinib , Lung Neoplasms , Ferroptosis/drug effects , Humans , Gefitinib/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/genetics , Cell Line, Tumor , Discoidin Domain Receptor 1/metabolism , Discoidin Domain Receptor 1/genetics , Cell Proliferation/drug effects , MicroRNAs/genetics , MicroRNAs/metabolism , Amino Acid Transport System y+/metabolism , Amino Acid Transport System y+/genetics , Gene Expression Regulation, Neoplastic/drug effects , Animals , Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Mice , Suppressor of Cytokine Signaling Proteins
SELECTION OF CITATIONS
SEARCH DETAIL