Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Biol Pharm Bull ; 47(7): 1288-1295, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39010214

RESUMEN

The active form of discoidin domain receptors (DDRs) is expressed in cell surface and regulated post-translationally by glucose. The DDR2 and DDR1 transfected in HEK293 cells were expressed mainly in their active forms with sizes of 130 and 120 kDa, respectively. DDRs were observed predominantly as 100 kDa proteins in glucose-depleted culture conditions. However, transfection of endothelial growth factor receptor (EGFR) in HEK293 cells resulted in the expression of only one form regardless of glucose concentration. Vascular smooth muscle cells, HT1080s, and MDA-MB-231 cancer cells expressed DDRs in their active forms in high glucose concentrations, which did not occur with EGFR. In diabetic rats, DDRs were expressed at high levels in arterial tissue but EGFR was not highly expressed. Taken together, these results suggest that DDRs expression depends on glucose concentration it may cooperate in the development of atherosclerosis and kidney fibroblasts, promoting nephropathy in diabetic rats.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Glucosa , Animales , Humanos , Glucosa/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/genética , Masculino , Diabetes Mellitus Experimental/metabolismo , Células HEK293 , Ratas , Arterias/metabolismo , Arterias/patología , Receptores ErbB/metabolismo , Receptores ErbB/genética , Línea Celular Tumoral , Receptor con Dominio Discoidina 2/metabolismo , Receptor con Dominio Discoidina 2/genética , Músculo Liso Vascular/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Ratas Wistar
2.
Mol Cancer Ther ; 23(8): 1124-1138, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38670553

RESUMEN

Oncogene-driven expression and activation of receptor tyrosine kinases promotes tumorigenesis and contributes to drug resistance. Increased expression of the kinases discoidin domain receptor 2 (DDR2), RET Proto-Oncogene (RET), Platelet Derived Growth Factor Receptor Alpha (PDGFRA), KIT Proto-Oncogene (KIT), MET Proto-Oncogene (MET), and anaplastic lymphoma kinase (ALK) independently correlate with decreased overall survival and event free survival of pediatric neuroblastoma. The multikinase inhibitor sitravatinib targets DDR2, RET, PDGFRA, KIT, and MET with low nanomolar activity and we therefore tested its efficacy against orthotopic and syngeneic tumor models. Sitravatinib markedly reduced cell proliferation and migration in vitro independently of N-Myc proto-oncogene (MYCN), ALK, or c-Myc proto-oncogene status and inhibited proliferation and metastasis of human orthotopic xenografts. Oral administration of sitravatinib to homozygous Th-MYCN transgenic mice (Th-MYCN+/+) after tumor initiation completely arrested further tumor development with no mice dying of disease while maintained on sitravatinib treatment (control cohort 57 days median time to sacrifice). Among these top kinases, DDR2 expression has the strongest correlation with poor survival and high stage at diagnosis and the highest sensitivity to the drug. We confirmed on-target inhibition of collagen-mediated activation of DDR2. Genetic knockdown of DDR2 partially phenocopies sitravatinib treatment, limiting tumor development and metastasis across tumor models. Analysis of single-cell sequencing data demonstrated that DDR2 is restricted to mesenchymal-type tumor subpopulations and is enriched in Schwann cell precursor subpopulations found in high-risk disease. These data define an unsuspected role for sitravatinib as a therapeutic agent in neuroblastoma and reveal a novel function for DDR2 as a driver of tumor growth and metastasis.


Asunto(s)
Receptor con Dominio Discoidina 2 , Neuroblastoma , Proto-Oncogenes Mas , Transducción de Señal , Neuroblastoma/patología , Neuroblastoma/metabolismo , Neuroblastoma/genética , Neuroblastoma/tratamiento farmacológico , Animales , Humanos , Ratones , Receptor con Dominio Discoidina 2/metabolismo , Receptor con Dominio Discoidina 2/genética , Metástasis de la Neoplasia , Proliferación Celular , Ensayos Antitumor por Modelo de Xenoinjerto , Línea Celular Tumoral , Ratones Transgénicos , Femenino
3.
FEBS Open Bio ; 14(5): 867-882, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38538106

RESUMEN

The extracellular matrix (ECM) regulates carcinogenesis by interacting with cancer cells via cell surface receptors. Discoidin Domain Receptor 2 (DDR2) is a collagen-activated receptor implicated in cell survival, growth, and differentiation. Dysregulated DDR2 expression has been identified in various cancer types, making it as a promising therapeutic target. Additionally, cancer cells exhibit mechanosensing abilities, detecting changes in ECM stiffness, which is particularly important for carcinogenesis given the observed ECM stiffening in numerous cancer types. Despite these, whether collagen-activated DDR2 signaling and ECM stiffness-induced mechanosensing exert similar effects on cancer cell behavior and whether they operate through analogous mechanisms remain elusive. To address these questions, we performed bulk RNA sequencing (RNA-seq) on human SH-SY5Y neuroblastoma cells cultured on collagen-coated substrates. Our results show that DDR2 downregulation induces significant changes in the cell transcriptome, with changes in expression of 15% of the genome, specifically affecting the genes associated with cell division and differentiation. We validated the RNA-seq results by showing that DDR2 knockdown redirects the cell fate from proliferation to senescence. Like DDR2 knockdown, increasing substrate stiffness diminishes cell proliferation. Surprisingly, RNA-seq indicates that substrate stiffness has no detectable effect on the transcriptome. Furthermore, DDR2 knockdown influences cellular responses to substrate stiffness changes, highlighting a crosstalk between these two ECM-induced signaling pathways. Based on our results, we propose that the ECM could activate DDR2 signaling and mechanosensing in cancer cells to orchestrate their cell fate through distinct mechanisms, with or without involving gene expression, thus providing novel mechanistic insights into cancer progression.


Asunto(s)
Receptor con Dominio Discoidina 2 , Neuroblastoma , Transducción de Señal , Transcriptoma , Humanos , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , Receptor con Dominio Discoidina 2/metabolismo , Receptor con Dominio Discoidina 2/genética , Transcriptoma/genética , Transducción de Señal/genética , Línea Celular Tumoral , Matriz Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Mecanotransducción Celular/genética , Diferenciación Celular/genética , Proliferación Celular/genética
4.
J Pathol ; 262(4): 505-516, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38332727

RESUMEN

Pulmonary fibrosis, especially idiopathic pulmonary fibrosis (IPF), portends significant morbidity and mortality, and current therapeutic options are suboptimal. We have previously shown that type I collagen signaling through discoidin domain receptor 2 (DDR2), a receptor tyrosine kinase expressed by fibroblasts, is critical for the regulation of fibroblast apoptosis and progressive fibrosis. However, the downstream signaling pathways for DDR2 remain poorly defined and could also be attractive potential targets for therapy. A recent phosphoproteomic approach indicated that PIK3C2α, a poorly studied member of the PI3 kinase family, could be a downstream mediator of DDR2 signaling. We hypothesized that collagen I/DDR2 signaling through PIK3C2α regulates fibroblast activity during progressive fibrosis. To test this hypothesis, we found that primary murine fibroblasts and IPF-derived fibroblasts stimulated with endogenous or exogenous type I collagen led to the formation of a DDR2/PIK3C2α complex, resulting in phosphorylation of PIK3C2α. Fibroblasts treated with an inhibitor of PIK3C2α or with deletion of PIK3C2α had fewer markers of activation after stimulation with TGFß and more apoptosis after stimulation with a Fas-activating antibody. Finally, mice with fibroblast-specific deletion of PIK3C2α had less fibrosis after bleomycin treatment than did littermate control mice with intact expression of PIK3Cα. Collectively, these data support the notion that collagen/DDR2/PIK3C2α signaling is critical for fibroblast function during progressive fibrosis, making this pathway a potential target for antifibrotic therapy. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Receptor con Dominio Discoidina 2 , Fibrosis Pulmonar Idiopática , Ratones , Animales , Receptor con Dominio Discoidina 2/genética , Receptor con Dominio Discoidina 2/metabolismo , Colágeno Tipo I/metabolismo , Fibroblastos/patología , Colágeno/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Receptores con Dominio Discoidina/metabolismo , Pulmón/patología
5.
Matrix Biol ; 125: 31-39, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38081526

RESUMEN

Discoidin domain receptors (DDRs), including DDR1 and DDR2, are a unique class of receptor tyrosine kinases (RTKs) activated by collagens at the cell-matrix boundary interface. The peculiar mode of activation makes DDRs as key cellular sensors of microenvironmental changes, with a critical role in all physiological and pathological processes governed by collagen remodeling. DDRs are widely expressed in fetal and adult tissues, and experimental and clinical evidence has shown that their expression is deregulated in cancer. Strong findings supporting the role of collagens in tumor progression and metastasis have led to renewed interest in DDRs.  However, despite an increasing number of studies, DDR biology remains poorly understood, particularly the less studied DDR2, whose involvement in cancer progression mechanisms is undoubted. Thus, the understanding of a wider range of DDR2 functions and related molecular mechanisms is expected. To date, several lines of evidence support DDR2 as a promising target in cancer therapy. Its involvement in key functions in the tumor microenvironment makes DDR2 inhibition particularly attractive to achieve simultaneous targeting of tumor and stromal cells, and tumor regression, which is beneficial for improving the response to different types of anti-cancer therapies, including chemo- and immunotherapy. This review summarizes current research on DDR2, focusing on its role in cancer progression through its involvement in tumor and stromal cell functions, and discusses findings that support the rationale for future development of direct clinical strategies targeting DDR2.


Asunto(s)
Receptor con Dominio Discoidina 2 , Neoplasias , Adulto , Humanos , Receptor con Dominio Discoidina 2/genética , Receptor con Dominio Discoidina 2/metabolismo , Receptores Mitogénicos/genética , Receptores Mitogénicos/metabolismo , Unión Proteica , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores con Dominio Discoidina/genética , Neoplasias/genética , Colágeno/metabolismo , Receptor con Dominio Discoidina 1/genética , Receptor con Dominio Discoidina 1/metabolismo , Microambiente Tumoral
6.
Pathol Res Pract ; 253: 155047, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38154356

RESUMEN

Ovarian cancer has the highest mortality among all gynecological malignancies. Therefore, it is urgent to determine the molecular mechanism of ovarian cancer progression. As the most prevalent modification of messenger RNA (mRNA), N6-Methyladenosine (m6A) modification is recognized as a key regulatory role in the progression of various tumors. However, the specific role of m6A and its related regulatory pathways in ovarian cancer (OV) remains unclear. In this study, we demonstrated that the METTL3/YTHDF1 m6A axis plays an important role in the progression of ovarian cancer. Depletion of METTL3/YTHDF1 impaired cancer proliferation and metastasis in vitro and in vivo. Mechanistically, The METTL3/YTHDF1 m6A axis directly binds to the mRNA of DDR2, thereby promoting the expression levels of the tumor promoter DDR2 and thus contributing to the progression of ovarian cancer. Collectively, our findings on the METTL3/YTHDF1/DDR2 m6A axis provide the insight into the underlying mechanism of ovarian carcinogenesis and highlight potential therapeutic targets for cancer treatment.


Asunto(s)
Receptor con Dominio Discoidina 2 , Neoplasias Ováricas , Humanos , Femenino , Carcinogénesis/genética , Transformación Celular Neoplásica , Carcinógenos , Neoplasias Ováricas/genética , ARN Mensajero/genética , Metiltransferasas/genética , Metiltransferasas/metabolismo , Proteínas de Unión al ARN/genética , Receptor con Dominio Discoidina 2/metabolismo
7.
Sheng Li Xue Bao ; 75(4): 515-520, 2023 Aug 25.
Artículo en Chino | MEDLINE | ID: mdl-37583038

RESUMEN

Pulmonary fibrosis is a severe lung interstitial disease characterized by the destruction of lung tissue structure, excessive activation and proliferation of fibroblasts, secretion and accumulation of a large amount of extracellular matrix (ECM), and impaired lung function. Due to the complexity of the disease, a suitable animal model to mimic human pulmonary fibrosis has not yet been established. Precision-cut lung slice (PCLS) has been a widely used in vitro method to study lung physiology and pathogenesis in recent years. This method is an in vitro culture technology at the level between organs and cells, because it can preserve the lung tissue structure and various types of airway cells in the lung tissue, simulate the in vivo lung environment, and conduct the observation of various interactions between cells and ECM. Therefore, PCLS can compensate for the limitations of other models such as cell culture. In order to explore the role of discoidin domain receptor 2 (DDR2) in pulmonary fibrosis, Ddr2flox/flox mice were successfully constructed. The Cre-LoxP system and PCLS technology were used to verify the deletion or knockdown of DDR2 in mouse PCLS. Transforming growth factor ß1 (TGF-ß1) can induce fibrosis of mouse PCLS in vitro, which can simulate the in vivo environment of pulmonary fibrosis. In the DDR2 knock down-PCLS in vitro model, the expression of various fibrosis-related factors induced by TGF-ß1 was significantly reduced, suggesting that knocking down DDR2 can inhibit the formation of pulmonary fibrosis. The results provide a new perspective for the clinical study of DDR2 as a therapeutic target in pulmonary fibrosis.


Asunto(s)
Receptor con Dominio Discoidina 2 , Fibrosis Pulmonar , Animales , Humanos , Ratones , Receptor con Dominio Discoidina 2/genética , Receptor con Dominio Discoidina 2/metabolismo , Fibroblastos/patología , Fibrosis , Pulmón/patología , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
8.
Mol Cancer Res ; 21(11): 1234-1248, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37527178

RESUMEN

Ovarian cancer is the leading cause of gynecologic cancer-related deaths. The propensity for metastasis within the peritoneal cavity is a driving factor for the poor outcomes associated with this disease, but there is currently no effective therapy targeting metastasis. In this study, we investigate the contribution of stromal cells to ovarian cancer metastasis and identify normal stromal cell expression of the collagen receptor, discoidin domain receptor 2 (DDR2), that acts to facilitate ovarian cancer metastasis. In vivo, global genetic inactivation of Ddr2 impairs the ability of Ddr2-expressing syngeneic ovarian cancer cells to spread throughout the peritoneal cavity. Specifically, DDR2 expression in mesothelial cells lining the peritoneal cavity facilitates tumor cell attachment and clearance. Subsequently, omentum fibroblast expression of DDR2 promotes tumor cell invasion. Mechanistically, we find DDR2-expressing fibroblasts are more energetically active, such that DDR2 regulates glycolysis through AKT/SNAI1 leading to suppressed fructose-1,6-bisphosphatase and increased hexokinase activity, a key glycolytic enzyme. Upon inhibition of DDR2, we find decreased protein synthesis and secretion. Consequently, when DDR2 is inhibited, there is reduction in secreted extracellular matrix proteins important for metastasis. Specifically, we find that fibroblast DDR2 inhibition leads to decreased secretion of the collagen crosslinker, LOXL2. Adding back LOXL2 to DDR2 deficient fibroblasts rescues the ability of tumor cells to invade. Overall, our results suggest that stromal cell expression of DDR2 is an important mediator of ovarian cancer metastasis. IMPLICATIONS: DDR2 is highly expressed by stromal cells in ovarian cancer that can mediate metastasis and is a potential therapeutic target in ovarian cancer.


Asunto(s)
Receptor con Dominio Discoidina 2 , Neoplasias Ováricas , Femenino , Humanos , Receptor con Dominio Discoidina 2/genética , Receptor con Dominio Discoidina 2/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Fosforilación , Colágeno/metabolismo , Matriz Extracelular/metabolismo
9.
Am J Physiol Lung Cell Mol Physiol ; 325(3): L342-L351, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37489274

RESUMEN

Progressive pulmonary fibrosis is a devastating condition and current treatment is suboptimal. There has been considerable interest in the role of tyrosine kinase signaling as mediators of pro- and antifibrotic processes. Nintedanib is a nonspecific tyrosine kinase that has been shown to have therapeutic benefit in lung fibrosis. However, the precise mechanism of action remains unclear because nintedanib inhibits several tyrosine kinases, which are often expressed on multiple cell types with different activities during fibrosis. Discoidin domain receptor 2 (DDR2) has been suggested as a potential target of nintedanib. DDR2 is a receptor tyrosine kinase that is activated by fibrillar collagens such as type I collagen. DDR2 is primarily expressed by fibroblasts. The effectiveness of specifically targeting DDR2 signaling during fibrosis remains undefined. In the present study, we show that nintedanib acts as a direct and indirect inhibitor of DDR2. We then utilize a novel allosteric inhibitor of DDR2, WRG-28, which blocks ligand binding and activation of DDR2. We find that WRG-28 augments fibroblast apoptosis and attenuates fibrosis. Finally, we show that fibroblast type I collagen autocrine signaling is regulated by DDR2 through both kinase-dependent and kinase-independent functions of DDR2. These findings highlight the importance of type I collagen autocrine signaling by fibroblasts during fibrosis and demonstrate that DDR2 has a central role in this pathway making it a potential therapeutic target.NEW & NOTEWORTHY Type I collagen is a major component of fibrosis and can signal through cell surface receptors such as discoidin domain receptor 2 (DDR2). DDR2 activation can lead to further collagen deposition by fibroblasts setting up a profibrotic positive feedback loop. In this report, we find that inhibition of DDR2 with nintedanib or a specific DDR2 inhibitor, WRG-28, can disrupt this cycle and prevent fibrosis through augmented fibroblast apoptosis and inhibited activation.


Asunto(s)
Receptor con Dominio Discoidina 2 , Humanos , Receptor con Dominio Discoidina 2/metabolismo , Colágeno Tipo I/metabolismo , Fibroblastos/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Fibrosis
10.
Elife ; 122023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-37405383

RESUMEN

Separate tissues connect through adjoining basement membranes to carry out molecular barrier, exchange, and organ support functions. Cell adhesion at these connections must be robust and balanced to withstand independent tissue movement. Yet, how cells achieve synchronized adhesion to connect tissues is unknown. Here, we have investigated this question using the Caenorhabditis elegans utse-seam tissue connection that supports the uterus during egg-laying. Through genetics, quantitative fluorescence, and cell-specific molecular disruption, we show that type IV collagen, which fastens the linkage, also activates the collagen receptor discoidin domain receptor-2 (DDR-2) in both the utse and seam. RNAi depletion, genome editing, and photobleaching experiments revealed that DDR-2 signals through LET-60/Ras to coordinately strengthen an integrin adhesion in the utse and seam that stabilizes their connection. These results uncover a synchronizing mechanism for robust adhesion during tissue connection, where collagen both affixes the linkage and signals to both tissues to bolster their adhesion.


Asunto(s)
Receptor con Dominio Discoidina 2 , Integrinas , Animales , Femenino , Receptores con Dominio Discoidina/metabolismo , Transducción de Señal , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Colágeno/metabolismo , Adhesión Celular/fisiología , Receptor con Dominio Discoidina 2/metabolismo
11.
Bioorg Chem ; 135: 106506, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37030105

RESUMEN

We report the synthesis, molecular docking and anticancer properties of the novel compound (E)-1-methyl-9-(3-methylbenzylidene)-6,7,8,9-tetrahydropyrazolo[3,4-d]pyrido[1,2-a]pyrimidin-4(1H)-one (PP562). PP562 was screened against sixteen human cancer cell lines and exhibited excellent antiproliferative activity with IC50 values ranging from 0.016 to 5.667 µM. Experiments were carried out using the target PP562 at a single dose of 1.0 µM against a kinase panel comprising 100 different enzymes. A plausible binding mechanism for PP562 inhibition of DDR2 was determined using molecular dynamic analysis. The effect of PP562 on cell proliferation was also examined in cancer cell models with both high and low expression of the DDR2 gene; PP562 inhibition of high-expressing cells was more prominent than that for low expressing cells. PP562 also exhibits excellent anticancer potency toward the HGC-27 gastric cancer cell line. In addition, PP562 inhibits colony formation, cell migration, and adhesion, induces cell cycle arrest at the G2/M phase, and affects ROS generation and cell apoptosis. After DDR2 gene knockdown, the antitumor effects of PP562 on tumor cells were significantly impaired. These results suggested that PP562 might exert its inhibitory effect on HCG-27 proliferation through the DDR2 target.


Asunto(s)
Antineoplásicos , Receptor con Dominio Discoidina 2 , Humanos , Simulación del Acoplamiento Molecular , Receptor con Dominio Discoidina 2/metabolismo , Línea Celular Tumoral , Antineoplásicos/química , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/química , Proliferación Celular , Estructura Molecular , Ensayos de Selección de Medicamentos Antitumorales , Relación Estructura-Actividad
12.
Cell Signal ; 106: 110636, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36813149

RESUMEN

BACKGROUND: Peritoneal metastasis (PM) is an independent prognostic factor in gastric cancer (GC), however, the underlying mechanisms of PM occurrence remain unclear. METHOD: The roles of DDR2 were investigated in GC and its potential relationship to PM, and orthotopic implants into nude mice were performed to assess the biological effects of DDR2 on PM. RESULTS: Herein, DDR2 level is more significantly observed to elevate in PM lesion than the primary lesion. GC with DDR2-high expression evokes a worse overall survival (OS) in TCGA, similar results of the gloomy OS with high DDR2 levels are clarified via the stratifying stage of TNM. The conspicuously increased expression of DDR2 was found in GC cell lines, luciferase reporter assays verified that miR-199a-3p directly targeted DDR2 gene, which was correlated to tumor progression. We ulteriorly observed DDR2 participated in GC stemness maintenance via mediating pluripotency factor SOX2 expression and implicated in autophagy and DNA damage of cancer stem cells (CSCs). In particular, DDR2 dominated EMT programming through recruiting NFATc1-SOX2 complex to Snai1 in governing cell progression, controlling by DDR2-mTOR-SOX2 axis in SGC-7901 CSCs. Furthermore, DDR2 promoted the tumor peritoneal dissemination in gastric xenograft mouse model. CONCLUSION: Phenotype screens and disseminated verifications incriminating in GC exposit the miR-199a-3p-DDR2-mTOR-SOX2 axis as a clinically actionable target for tumor PM progression. The herein-reported DDR2-based underlying axis in GC represents novel and potent tools for studying the mechanisms of PM.


Asunto(s)
Receptor con Dominio Discoidina 2 , MicroARNs , Neoplasias Gástricas , Animales , Humanos , Ratones , Línea Celular Tumoral , Proliferación Celular/genética , Receptor con Dominio Discoidina 2/genética , Receptor con Dominio Discoidina 2/metabolismo , Regulación Neoplásica de la Expresión Génica , Ratones Desnudos , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias Gástricas/patología , Serina-Treonina Quinasas TOR/metabolismo , Células Madre Neoplásicas
13.
Am J Physiol Lung Cell Mol Physiol ; 324(3): L307-L324, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36719983

RESUMEN

The extracellular matrix (ECM) of the pulmonary parenchyma must maintain the structural relationships among resident cells during the constant distortion imposed by respiration. This dictates that both the ECM and cells adapt to changes in shape, while retaining their attachment. Membrane-associated integrins and discoidin domain receptors (DDR) bind collagen and transmit signals to the cellular cytoskeleton. Although the contributions of DDR2 to collagen deposition and remodeling during osseous development are evident, it is unclear how DDR2 contributes to lung development. Using mice (smallie, Slie/Slie, DDR2Δ) bearing a spontaneous inactivating deletion within the DDR2 coding region, we observed a decrease in gas-exchange surface area and enlargement of alveolar ducts. Compared with fibroblasts isolated from littermate controls, DDR2Δ fibroblasts, spread more slowly, developed fewer lamellipodia, and were less responsive to the rigidity of neighboring collagen fibers. Activated ß1-integrin (CD29) was reduced in focal adhesions (FA) of DDR2Δ fibroblasts, less phospho-zyxin localized to and fewer FA developed over ventral actin stress fibers, and the adhesions had a lower aspect ratio compared with controls. However, DDR2 deletion did not reduce cellular displacement of the ECM. Our findings indicate that DDR2, in concert with collagen-binding ß1-integrins, regulates the timing and location of focal adhesion formation and how lung fibroblasts respond to ECM rigidity. Reduced rigidity sensing and mechano-responsiveness may contribute to the distortion of alveolar ducts, where the fiber cable-network is enriched and tensile forces are concentrated. Strategies targeting DDR2 could help guide fibroblasts to locations where tensile forces organize parenchymal repair.


Asunto(s)
Receptor con Dominio Discoidina 2 , Ratones , Animales , Receptor con Dominio Discoidina 2/metabolismo , Integrinas/metabolismo , Adhesiones Focales/metabolismo , Receptores con Dominio Discoidina , Colágeno/metabolismo , Pulmón/metabolismo
14.
Med Mol Morphol ; 56(1): 46-57, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36370214

RESUMEN

The aim of the study was to correlate the immunohistochemical expression of cartilage intermediate layer protein 2 (CILP-2) and discoidin domain receptor 2 (DDR2), and the ultrastructural changes in the cartilage with the degree of articular cartilage damage in osteoarthritis (OA) patients. Cartilage samples were obtained from twenty patients aged from 46 to 68 years undergoing total knee arthroplasty. In each patient, medial and lateral tibial plateau samples were analysed applying OARSI histopathology grading. Positive correlation was noted between the extent of CILP-2 staining intensity and OARSI grades. Abundant staining for CILP-2 was found in the superficial and middle layers and in the pericellular matrix (PCM) of the deep zone. Transmission electron microscopy studies demonstrated strong damage of chondrocytes, the organelles were often diminished or focally aggregated. As a characteristic finding, PCM was frequently expanded, which may reflect a pathogenic step in OA progression. In conclusion, CILP-2 may potentially be a relevant marker of OA progression as its expression correlated better with cartilage damage than the known marker of articular cartilage damage, DDR2.


Asunto(s)
Artroplastia de Reemplazo de Rodilla , Cartílago Articular , Receptor con Dominio Discoidina 2 , Osteoartritis de la Rodilla , Humanos , Persona de Mediana Edad , Anciano , Osteoartritis de la Rodilla/metabolismo , Osteoartritis de la Rodilla/patología , Cartílago Articular/patología , Receptor con Dominio Discoidina 2/metabolismo , Articulación de la Rodilla , Condrocitos , Pirofosfatasas/metabolismo , Proteínas de la Matriz Extracelular/metabolismo
15.
Cytokine ; 158: 155981, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35952595

RESUMEN

BACKGROUND: Adipose-derived stem cells (ADSCs) and their derived exosomes (ADSC-Exos) have shown potential functions in tissue repair. This study focuses on the effects of ADSCs-Exos on cutaneous wound healing and the potential involvement of the long non-coding RNA (lncRNA) XIST/microRNA-96-5p (miR-96-5p)/discoidin domain receptor 2 (DDR2) axis. METHODS: Exos were isolated from the ADSCs and identified. A mouse model of full-thickness skin wounds was established. The mice were treated with ADSC-Exos to evaluate the function of ADSC-Exos in wound healing. Mouse dermal fibroblasts (MDFs) were co-cultured with the ADSC-Exos for in vitro experiments. The most differentially expressed lncRNAs in mouse skin tissues after ADSC-Exo treatment were screened by microarray analysis. The downstream molecules were analyzed by bioinformatics tools. Gain- and loss-of-function studies were performed to examine the functions of the XIST/miR-96-5p/DDR2 axis in wound healing. RESULTS: ADSC-Exos facilitated wound healing in mice, reduced inflammatory infiltration, and increased collagen deposition in the wound skin tissues. In vitro, the ADSC-Exos promoted proliferation, migration of the MDFs. XIST was the most upregulated lncRNA in MDFs after ADSC-Exo treatment. Downregulation of XIST suppressed the promoting role of ADSC-Exos in wound healing. XIST bound to miR-96-5p to restore the expression of DDR2 mRNA. Either silencing of miR-96-5p or overexpression of DDR2 restored the promoting functions of ADSC-Exos in proliferation and migration of MDFs. CONCLUSION: This study demonstrates that ADSC-Exos-carried XIST accelerates cutaneous wound healing through suppressing miR-96-5p and restoring the DDR2 expression.


Asunto(s)
Receptor con Dominio Discoidina 2/metabolismo , Exosomas , MicroARNs , ARN Largo no Codificante/genética , Animales , Exosomas/metabolismo , Ratones , MicroARNs/genética , MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Células Madre , Cicatrización de Heridas/genética
16.
Biomater Sci ; 10(16): 4596-4611, 2022 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-35792605

RESUMEN

RNA interference is a promising way to treat cancer and the construction of a stable drug delivery system is critically important for its application. Gelatin nanospheres (GNs) comprise a biodegradable drug vehicle with excellent biocompatibility, but there are limited studies on its delivery and role in the stabilization of miRNA and siRNA. Breast cancer is the most diagnosed type of female cancer worldwide. Abnormal miRNA expression is closely related to the occurrence and progression of estrogen receptor-positive (ER+) breast cancer. In this study, miR-4458 was upregulated in ER+ breast cancer and could inhibit MCF-7 cell viability, colony formation, migration, and invasion. Collagen type XI alpha 1 (COL11A1) was identified as a directly interacting protein of miR-4458 and an important component of the extracellular matrix. High COL11A1 expression was positively correlated with poor prognosis, lower overall survival, disease-free survival, and a late tumor-node-metastasis stage. COL11A1 knockdown could inhibit MCF-7 cell migration and invasion. GNs were used to load a miR-4458 mimic or COL11A1 siRNA (si-COL11A1) to achieve sustained and controlled release in xenograft nude mice. Their tumor volume was decreased, tumor cell apoptosis was promoted, and hepatic metastasis was significantly inhibited. Moreover, the DDR2/SRC signaling pathway was inactivated after transfection with the miR-4458 mimic and si-COL11A1. In conclusion, GNs can be potentially used to deliver siRNA or miRNA, and miR-4458 and COL11A1 can be possible targets for ER+ breast cancer treatment.


Asunto(s)
Neoplasias de la Mama , Receptor con Dominio Discoidina 2 , MicroARNs , Nanosferas , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular , Colágeno Tipo XI/genética , Colágeno Tipo XI/metabolismo , Receptor con Dominio Discoidina 2/genética , Receptor con Dominio Discoidina 2/metabolismo , Femenino , Gelatina/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Desnudos , MicroARNs/metabolismo , ARN Interferente Pequeño/uso terapéutico , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Receptores de Estrógenos/uso terapéutico , Transducción de Señal , Familia-src Quinasas
17.
Breast J ; 2022: 5169405, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35711892

RESUMEN

Discoidin domain receptor 2 (DDR2) is arising as a promising therapeutic target in breast carcinoma (BC). The ability of DDR2 to bind to collagen promotes protumoral responses in cancer cells that influence the tumor microenvironment (TME). Nonetheless, the interrelation between DDR2 expression and TME modulation during BC progression remains poorly known. For this reason, we aim to evaluate the correlation between intratumoral expression of DDR2 and the infiltration of the main TME cell populations, cancer-associated fibroblasts (CAFs), and tumor-associated macrophages (TAMs). First, collagen and DDR2 expression levels were analyzed in human invasive BC samples. Then, DDR2 status correlation with tumor aggressiveness and patient survival were retrieved from different databases. Subsequently, the main pathways, cell types, and tissues correlated with DDR2 expression in BC were obtained through bioinformatics approach. Finally, we studied the association of DDR2 expression with the recruitment of CAFs and TAMs. Our findings showed that, together with the expected overexpression of TME markers, DDR2 was upregulated in tumor samples. Besides, we uncovered that altered TME markers were linked to DDR2 expression in invasive BC patients. Consequently, DDR2 modulates the stromal reaction through CAFs and TAMs infiltration and could be used as a potential worse prognostic factor in the treatment response of invasive BC.


Asunto(s)
Neoplasias de la Mama , Fibroblastos Asociados al Cáncer , Receptor con Dominio Discoidina 2 , Macrófagos Asociados a Tumores , Neoplasias de la Mama/patología , Colágeno/metabolismo , Receptor con Dominio Discoidina 2/genética , Receptor con Dominio Discoidina 2/metabolismo , Femenino , Humanos , Pronóstico , Microambiente Tumoral
18.
J Cancer Res Clin Oncol ; 148(12): 3385-3398, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35761108

RESUMEN

PURPOSE: To investigate the roles of DDR2 and IFITM1 in breast cancer (BC). METHODS: The expression of DDR2 and IFITM1 in BC tissues and cell lines was measured. DDR2 and/or IFITM1 were knocked down in BT20 and MDA-MB-231 cells, after which the viability, mobility and apoptosis of the cells were tested. Xenograft mouse models were established through subcutaneous tumor transplantation. RESULTS: DDR2 and IFITM1 were highly expressed in invasive BC tissues and cell lines. Overexpression of DDR2 and/or IFITM1 was associated with poorer clinical outcomes and patient survival. Knockdown of DDR2 or IFITM1 suppressed the viability and invasiveness of BT20 and MDA-MB-231 cells and restrained the growth of xenograft tumors in nude mice. Simultaneous knockdown of IFITM1 and DDR2 surpassed knockdown of IFITM1 alone in suppressing BC development. CONCLUSIONS: DDR2 and IFITM1 are co-expressed to facilitate the malignant behaviors of BC cells and promote the development of tumors.


Asunto(s)
Neoplasias de la Mama , Receptor con Dominio Discoidina 2 , Humanos , Ratones , Animales , Femenino , Ratones Desnudos , Proliferación Celular/genética , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Apoptosis/genética , Neoplasias de la Mama/patología , Movimiento Celular/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Receptor con Dominio Discoidina 2/metabolismo
19.
Br J Pharmacol ; 179(19): 4709-4721, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35751904

RESUMEN

BACKGROUND AND PURPOSE: Tyrosine kinase inhibitors (TKI) used to treat chronic myeloid leukaemia (CML) have been associated with cardiovascular side effects, including reports of calcific aortic valve stenosis. The aim of this study was to establish the effects of first and second generation TKIs in aortic valve stenosis and to determine the associated molecular mechanisms. EXPERIMENTAL APPROACH: Hyperlipidemic APOE*3Leiden.CETP transgenic mice were treated with nilotinib, imatinib or vehicle. Human valvular interstitial cells (VICs) were isolated and studied in vitro. Gene expression analysis was perfromed in aortic valves from 64 patients undergoing aortic valve replacement surgery. KEY RESULTS: Nilotinib increased murine aortic valve thickness. Nilotinib, but not imatinib, promoted calcification and osteogenic activation and decreased autophagy in human VICs. Differential tyrosine kinase expression was detected between healthy and calcified valve tissue. Transcriptomic target identification revealed that the discoidin domain receptor DDR2, which is preferentially inhibited by nilotinib, was predominantly expressed in human aortic valves but markedly downregulated in calcified valve tissue. Nilotinib and selective DDR2 targeting in VICs induced a similar osteogenic activation, which was blunted by increasing the DDR2 ligand, collagen. CONCLUSIONS AND IMPLICATIONS: These findings suggest that inhibition of DDR2 by nilotinib promoted aortic valve thickening and VIC calcification, with possible translational implications for cardiovascular surveillance and possible personalized medicine in CML patients.


Asunto(s)
Estenosis de la Válvula Aórtica , Calcinosis , Receptor con Dominio Discoidina 2 , Animales , Válvula Aórtica/metabolismo , Válvula Aórtica/patología , Estenosis de la Válvula Aórtica/tratamiento farmacológico , Estenosis de la Válvula Aórtica/genética , Estenosis de la Válvula Aórtica/metabolismo , Calcinosis/tratamiento farmacológico , Calcinosis/genética , Calcinosis/metabolismo , Células Cultivadas , Receptor con Dominio Discoidina 2/metabolismo , Receptores con Dominio Discoidina/metabolismo , Humanos , Mesilato de Imatinib , Ratones , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas
20.
Cell Death Differ ; 29(4): 737-749, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34645939

RESUMEN

Obesity is closely associated with low-bone-mass disorder. Discoidin domain receptor 2 (DDR2) plays essential roles in skeletal metabolism, and is probably involved in fat metabolism. To test the potential role of DDR2 in fat and fat-bone crosstalk, Ddr2 conditional knockout mice (Ddr2Adipo) were generated in which Ddr2 gene is exclusively deleted in adipocytes by Adipoq Cre. We found that Ddr2Adipo mice are protected from fat gain on high-fat diet, with significantly decreased adipocyte size. Ddr2Adipo mice exhibit significantly increased bone mass and mechanical properties, with enhanced osteoblastogenesis and osteoclastogenesis. Marrow adipocyte is diminished in the bone marrow of Ddr2Adipo mice, due to activation of lipolysis. Fatty acid in the bone marrow was reduced in Ddr2Adipo mice. RNA-Seq analysis identified adenylate cyclase 5 (Adcy5) as downstream molecule of Ddr2. Mechanically, adipocytic Ddr2 modulates Adcy5-cAMP-PKA signaling, and Ddr2 deficiency stimulates lipolysis and supplies fatty acid for oxidation in osteoblasts, leading to the enhanced osteoblast differentiation and bone mass. Treatment of Adcy5 specific inhibitor abolishes the increased bone mass gain in Ddr2Adipo mice. These observations establish, for the first time, that Ddr2 plays an essential role in the crosstalk between fat and bone. Targeting adipocytic Ddr2 may be a potential strategy for treating obesity and pathological bone loss simultaneously.


Asunto(s)
Adipocitos , Densidad Ósea , Receptor con Dominio Discoidina 2 , Adipocitos/metabolismo , Animales , Receptor con Dominio Discoidina 2/metabolismo , Ácidos Grasos/metabolismo , Ratones , Obesidad/genética , Obesidad/metabolismo , Osteoblastos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...