Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
J Pathol ; 262(4): 505-516, 2024 04.
Article in English | MEDLINE | ID: mdl-38332727

ABSTRACT

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.


Subject(s)
Discoidin Domain Receptor 2 , Idiopathic Pulmonary Fibrosis , Mice , Animals , Discoidin Domain Receptor 2/genetics , Discoidin Domain Receptor 2/metabolism , Collagen Type I/metabolism , Fibroblasts/pathology , Collagen/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Discoidin Domain Receptors/metabolism , Lung/pathology
2.
DNA Repair (Amst) ; 134: 103626, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38232606

ABSTRACT

DNA double-strand breaks initiate the DNA damage response (DDR), leading to the accumulation of repair proteins at break sites and the formation of the-so-called foci. Various microscopy methods, such as wide-field, confocal, electron, and super-resolution microscopy, have been used to study these structures. However, the impact of different DNA-damaging agents on their (nano)structure remains unclear. Utilising GSDIM super-resolution microscopy, here we investigated the distribution of fluorescently tagged DDR proteins (53BP1, RNF168, MDC1) and γH2AX in U2OS cells treated with γ-irradiation, etoposide, cisplatin, or hydroxyurea. Our results revealed that both foci structure and their nanoscale ultrastructure, including foci size, nanocluster characteristics, fluorophore density and localisation, can be significantly altered by different inducing agents, even ones with similar mechanisms. Furthermore, distinct behaviours of DDR proteins were observed under the same treatment. These findings have implications for cancer treatment strategies involving these agents and provide insights into the nanoscale organisation of the DDR.


Subject(s)
DNA Repair , Microscopy , DNA Damage , Tumor Suppressor p53-Binding Protein 1/metabolism , DNA , Discoidin Domain Receptors/genetics , Discoidin Domain Receptors/metabolism
3.
Biomolecules ; 13(10)2023 10 13.
Article in English | MEDLINE | ID: mdl-37892200

ABSTRACT

Exposure to high acute doses of ionizing radiation (IR) can induce cutaneous radiation syndrome. Weeks after such radiation insults, keratinocyte nuclei of the epidermis exhibit persisting genomic lesions that present as focal accumulations of DNA double-strand break (DSB) damage marker proteins. Knowledge about the nanostructure of these genomic lesions is scarce. Here, we compared the chromatin nano-architecture with respect to DNA damage response (DDR) factors in persistent genomic DNA damage regions and healthy chromatin in epidermis sections of two minipigs 28 days after lumbar irradiation with ~50 Gy γ-rays, using single-molecule localization microscopy (SMLM) combined with geometric and topological mathematical analyses. SMLM analysis of fluorochrome-stained paraffin sections revealed, within keratinocyte nuclei with perisitent DNA damage, the nano-arrangements of pATM, 53BP1 and Mre11 DDR proteins in γ-H2AX-positive focal chromatin areas (termed macro-foci). It was found that persistent macro-foci contained on average ~70% of 53BP1, ~23% of MRE11 and ~25% of pATM single molecule signals of a nucleus. MRE11 and pATM fluorescent tags were organized in focal nanoclusters peaking at about 40 nm diameter, while 53BP1 tags formed nanoclusters that made up super-foci of about 300 nm in size. Relative to undamaged nuclear chromatin, the enrichment of DDR protein signal tags in γ-H2AX macro-foci was on average 8.7-fold (±3) for 53BP1, 3.4-fold (±1.3) for MRE11 and 3.6-fold (±1.8) for pATM. The persistent macro-foci of minipig epidermis displayed a ~2-fold enrichment of DDR proteins, relative to DSB foci of lymphoblastoid control cells 30 min after 0.5 Gy X-ray exposure. A lasting accumulation of damage signaling and sensing molecules such as pATM and 53BP1, as well as the DSB end-processing protein MRE11 in the persistent macro-foci suggests the presence of diverse DNA damages which pose an insurmountable problem for DSB repair.


Subject(s)
DNA Repair , Histones , Animals , Swine , Swine, Miniature/genetics , Swine, Miniature/metabolism , Histones/metabolism , Dose-Response Relationship, Radiation , DNA Damage , Chromatin , Epidermis/metabolism , Discoidin Domain Receptors/genetics , Discoidin Domain Receptors/metabolism
4.
Elife ; 122023 07 05.
Article in English | MEDLINE | ID: mdl-37405383

ABSTRACT

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.


Subject(s)
Discoidin Domain Receptor 2 , Integrins , Animals , Female , Discoidin Domain Receptors/metabolism , Signal Transduction , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Collagen/metabolism , Cell Adhesion/physiology , Discoidin Domain Receptor 2/metabolism
5.
Int J Mol Sci ; 24(12)2023 Jun 11.
Article in English | MEDLINE | ID: mdl-37373151

ABSTRACT

The collagen family contains 28 proteins, predominantly expressed in the extracellular matrix (ECM) and characterized by a triple-helix structure. Collagens undergo several maturation steps, including post-translational modifications (PTMs) and cross-linking. These proteins are associated with multiple diseases, the most pronounced of which are fibrosis and bone diseases. This review focuses on the most abundant ECM protein highly implicated in disease, type I collagen (collagen I), in particular on its predominant chain collagen type I alpha 1 (COLα1 (I)). An overview of the regulators of COLα1 (I) and COLα1 (I) interactors is presented. Manuscripts were retrieved searching PubMed, using specific keywords related to COLα1 (I). COL1A1 regulators at the epigenetic, transcriptional, post-transcriptional and post-translational levels include DNA Methyl Transferases (DNMTs), Tumour Growth Factor ß (TGFß), Terminal Nucleotidyltransferase 5A (TENT5A) and Bone Morphogenic Protein 1 (BMP1), respectively. COLα1 (I) interacts with a variety of cell receptors including integrinß, Endo180 and Discoidin Domain Receptors (DDRs). Collectively, even though multiple factors have been identified in association to COLα1 (I) function, the implicated pathways frequently remain unclear, underscoring the need for a more spherical analysis considering all molecular levels simultaneously.


Subject(s)
Collagen Type I , Collagen , Collagen/metabolism , Collagen Type I/metabolism , Extracellular Matrix/metabolism , Discoidin Domain Receptors/metabolism , Receptors, Cell Surface/metabolism
6.
J Virol ; 97(5): e0046123, 2023 05 31.
Article in English | MEDLINE | ID: mdl-37098896

ABSTRACT

Parvoviruses are single-stranded DNA viruses that utilize host proteins to vigorously replicate in the nuclei of host cells, leading to cell cycle arrest. The autonomous parvovirus, minute virus of mice (MVM), forms viral replication centers in the nucleus which are adjacent to cellular DNA damage response (DDR) sites, many of which are fragile genomic regions prone to undergoing DDR during the S phase. Since the cellular DDR machinery has evolved to transcriptionally suppress the host epigenome to maintain genomic fidelity, the successful expression and replication of MVM genomes at these cellular sites suggest that MVM interacts with DDR machinery distinctly. Here, we show that efficient replication of MVM requires binding of the host DNA repair protein MRE11 in a manner that is independent of the MRE11-RAD50-NBS1 (MRN) complex. MRE11 binds to the replicating MVM genome at the P4 promoter, remaining distinct from RAD50 and NBS1, which associate with cellular DNA break sites to generate DDR signals in the host genome. Ectopic expression of wild-type MRE11 in CRISPR knockout cells rescues virus replication, revealing a dependence on MRE11 for efficient MVM replication. Our findings suggest a new model utilized by autonomous parvoviruses to usurp local DDR proteins that are crucial for viral pathogenesis and distinct from those of dependoparvoviruses, like adeno-associated virus (AAV), which require a coinfected helper virus to inactivate the local host DDR. IMPORTANCE The cellular DNA damage response (DDR) machinery protects the host genome from the deleterious consequences of DNA breaks and recognizes invading viral pathogens. DNA viruses that replicate in the nucleus have evolved distinct strategies to evade or usurp these DDR proteins. We have discovered that the autonomous parvovirus, MVM, which is used to target cancer cells as an oncolytic agent, depends on the initial DDR sensor protein MRE11 to express and replicate efficiently in host cells. Our studies reveal that the host DDR interacts with replicating MVM molecules in ways that are distinct from viral genomes being recognized as simple broken DNA molecules. These findings suggest that autonomous parvoviruses have evolved distinct mechanisms to usurp DDR proteins, which can be used to design potent DDR-dependent oncolytic agents.


Subject(s)
MRE11 Homologue Protein , Minute Virus of Mice , Parvoviridae Infections , Animals , Mice , Cell Cycle Proteins/metabolism , Discoidin Domain Receptors/genetics , Discoidin Domain Receptors/metabolism , DNA Damage , DNA Replication , Minute Virus of Mice/genetics , Parvoviridae Infections/genetics , Virus Replication/physiology , MRE11 Homologue Protein/metabolism
7.
Clin Res Hepatol Gastroenterol ; 47(3): 102091, 2023 03.
Article in English | MEDLINE | ID: mdl-36738855

ABSTRACT

OBJECTIVE: DNA damage repair (DDR) gene mutations gained interest in the treatment of metastatic pancreatic cancer (PC) patients, but their relevance in adjuvant setting is not well characterized. We assessed the prognostic and predictive potential of tumoral expression of DDR proteins along with clinical and tumor characteristics in patients with resected PC. PATIENTS AND METHODS: Patients with PC who underwent pancreatic resection in our institution between 2005 and 2017 were retrospectively retrieved. Tumoral expression of a panel of DDR proteins including BRCA1, BRCA2, ATM, and p53 with immunohistochemistry was evaluated and association with patient and tumor features as well as prognosis was assessed. RESULTS: 130 patients were included in the study. The median age was 61 and 66% were males, 57% had lymph node involvement and 17% had a vascular invasion. 25 patients (19%) had thrombosis at the time of diagnosis. Median overall survival (OS) and disease-free survival (DFS) were 21.6 and 11.8 months, respectively. More advanced disease stage (HR: 3.67 95% CI 1.48-9.12, p = 0.005), presence of thrombosis (HR: 2.01 95% CI 1.04-3.89, p = 0.039), high BRCA1 expression (HR: 2.25, 95% CI 1.13-5.48, p = 0.023) and high post-operative CA 19-9 level (>100 IU/ml) (HR:2.61 95% CI 1.40-4.89, p = 0.003) were associated with shorter DFS. BRCA2, ATM, and p53 expression were not associated with DFS or OS. Adjuvant gemcitabine-cisplatin regimen was not associated with increased DFS or OS in the whole group, neither in low or high expressors of BRCA1, BRCA2, ATM or p53. CONCLUSION: Contrary to BRCA2, ATM, and P53, BRCA1 expression may be beneficial for prognosis in resected pancreatic cancer, while no predictive role was observed in terms of adjuvant platinum efficacy.


Subject(s)
Pancreatic Neoplasms , Tumor Suppressor Protein p53 , Male , Humans , Middle Aged , Female , Prognosis , Retrospective Studies , Tumor Suppressor Protein p53/genetics , Pancreatic Neoplasms/pathology , DNA Damage , Discoidin Domain Receptors/genetics , Discoidin Domain Receptors/metabolism , Pancreatic Neoplasms
8.
FEBS Lett ; 596(21): 2795-2807, 2022 11.
Article in English | MEDLINE | ID: mdl-36266942

ABSTRACT

The ErbB2 receptor tyrosine kinase plays a key role in mammary gland development. It forms large clusters which serve as signaling platforms for integration of extracellular information. The discoidin domain receptor (DDR) family are collagen receptor tyrosine kinases which, together with ErbB2, are involved in many physiological and pathological processes. Here, we investigated the interaction of ErbB2 and DDR1 receptors in breast cancer cells. In contrast to beta1-integrin, DDR1 colocalizes with ErbB2 in membrane clusters regardless of their expression levels. We demonstrated that this spatial coexistence is a consequence of the physical interaction between these receptors. In addition, these receptors are coexpressed in the normal mammary gland but not in breast tumor samples. Together, these results present DDR1 as a novel modulator of the ErbB2/ErbB3 signaling pathway.


Subject(s)
Discoidin Domain Receptor 1 , Receptor Protein-Tyrosine Kinases , Discoidin Domain Receptor 1/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Discoidin Domain Receptors/metabolism , Epithelial Cells/metabolism
9.
Phytomedicine ; 104: 154181, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35792445

ABSTRACT

BACKGROUND: Accumulation of age-associated senescent cells accompanied with increased reactive oxygen species (ROS) and inflammatory factors contributes to the progression of age-related macular degeneration (AMD), the main cause of blindness in the elderly. Berberine (BBR) has shown efficacy in the treatment of age-related diseases including diabetes and obesity by decreasing ROS. However, the pharmacological effect of BBR on alleviating retinal aging remains largely unknown. PURPOSE: Our study aimed to investigate the pharmacological effect of BBR as an anti-aging agent in retinal aging and its further molecular mechanisms. METHODS: D-galactose (DG)-induced ARPE-19 cell senescence and retinal aging were employed to evaluate the anti-aging effect of BBR in vivo and in vitro. The siRNA transfection, Western-Blot analyses, SA-ß-Gal assay and immunofluorescence were performed to investigate the potential mechanisms of BBR on anti-aging of RPE. RESULTS: In RPE-choroid of both natural aged and DG-induced accelerated aged mice, oxidative stress was increased along with the up-regulation of p21 expression, which was ameliorated by BBR treatment. BBR down-regulated the expression of REDD1 to decrease intracellular ROS content, attenuating DG-induced senescence in vitro and in vivo. Furthermore, p53 instead of HIF-1α was identified as the transcriptional regulator of REDD1 in DG-induced premature senescence. Importantly, NAC and BBR reversed the expression of p53 and the content of 8-OHdG, indicating that the positive feedback loop of ROS-DNA damage response (DDR) was formed, and BBR interrupted this feedback loop to alleviate DG-induced premature senescence by reducing REDD1 expression. In addition, BBR restored DG-damaged autophagy flux by up-regulating TFEB-mediated lysosomal biosynthesis by inhibiting REDD1 expression, thereby attenuating cellular senescence. CONCLUSION: BBR down-regulates REDD1 expression to interrupt the ROS-DDR positive feedback loop and restore autophagic flux, thereby reducing premature senescence of RPE. Our findings elucidate the promising effects of REDD1 on cellular senescence and the great potential of BBR as a therapeutic approach.


Subject(s)
Berberine , Retinal Pigment Epithelium , Transcription Factors/metabolism , Animals , Berberine/pharmacology , Cellular Senescence , Discoidin Domain Receptors/metabolism , Down-Regulation , Feedback , Mice , Oxidative Stress , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism
10.
Br J Pharmacol ; 179(19): 4709-4721, 2022 10.
Article in English | MEDLINE | ID: mdl-35751904

ABSTRACT

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.


Subject(s)
Aortic Valve Stenosis , Calcinosis , Discoidin Domain Receptor 2 , Animals , Aortic Valve/metabolism , Aortic Valve/pathology , Aortic Valve Stenosis/drug therapy , Aortic Valve Stenosis/genetics , Aortic Valve Stenosis/metabolism , Calcinosis/drug therapy , Calcinosis/genetics , Calcinosis/metabolism , Cells, Cultured , Discoidin Domain Receptor 2/metabolism , Discoidin Domain Receptors/metabolism , Humans , Imatinib Mesylate , Mice , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrimidines
11.
Acta Biochim Biophys Sin (Shanghai) ; 54(5): 615-624, 2022 May 25.
Article in English | MEDLINE | ID: mdl-35593476

ABSTRACT

Cellular energy metabolism dysregulation is associated with colorectal cancer (CRC) development and progression. Discoidin domain receptor 1a (DDR1a), one of the five DDR1 isoforms, is closely related to cell proliferation, invasion, and apoptosis in various tumors. Whether it participates in cellular metabolic reprogramming and regulates CRC initiation and progression remains unclear. In this study, we compared the expression of DDR1 in CRC tissues and adjacent tissues from 126 postoperative CRC samples. Moreover, lentivirus-mediated DDR1a overexpression and knockdown were performed in LoVo cells, and cell viability and proliferation were determined by CCK-8 and BrdU assays, respectively. Oxygen consumption rate, extracellular acidification rate, and lactate production were used to determine the effect of DDR1a on metabolic reprogramming. Clinically, CRC patients with high DDR1 expression had poor differentiation and were at an advanced TNM stage. DDR1a promoted LoVo cell proliferation, mitochondrial function, and extracellular acidification. Moreover, DDR1a knockdown inhibited intracellular lactic acid production in LoVo cells, while a pyruvate kinase inhibitor (diamide, 200 µM) significantly reversed this progression. Taken together, our results reveal that DDR1 plays a crucial role in maintaining intracellular environment homeostasis through metabolic reprogramming.


Subject(s)
Cell Proliferation , Colorectal Neoplasms , Discoidin Domain Receptor 1 , Energy Metabolism , Humans , Bromodeoxyuridine/metabolism , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Diamide , Discoidin Domain Receptor 1/genetics , Discoidin Domain Receptor 1/metabolism , Discoidin Domain Receptors/metabolism , Energy Metabolism/genetics , Lactic Acid , Protein Isoforms/metabolism , Pyruvate Kinase/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Sincalide/metabolism
12.
Hepatology ; 76(5): 1376-1388, 2022 11.
Article in English | MEDLINE | ID: mdl-35313030

ABSTRACT

BACKGROUND AND AIMS: Resolution of pathways that converge to induce deleterious effects in hepatic diseases, such as in the later stages, have potential antifibrotic effects that may improve outcomes. We aimed to explore whether humans and rodents display similar fibrotic signaling networks. APPROACH AND RESULTS: We assiduously mapped kinase pathways using 340 substrate targets, upstream bioinformatic analysis of kinase pathways, and over 2000 random sampling iterations using the PamGene PamStation kinome microarray chip technology. Using this technology, we characterized a large number of kinases with altered activity in liver fibrosis of both species. Gene expression and immunostaining analyses validated many of these kinases as bona fide signaling events. Surprisingly, the insulin receptor emerged as a considerable protein tyrosine kinase that is hyperactive in fibrotic liver disease in humans and rodents. Discoidin domain receptor tyrosine kinase, activated by collagen that increases during fibrosis, was another hyperactive protein tyrosine kinase in humans and rodents with fibrosis. The serine/threonine kinases found to be the most active in fibrosis were dystrophy type 1 protein kinase and members of the protein kinase family of kinases. We compared the fibrotic events over four models: humans with cirrhosis and three murine models with differing levels of fibrosis, including two models of fatty liver disease with emerging fibrosis. The data demonstrate a high concordance between human and rodent hepatic kinome signaling that focalizes, as shown by our network analysis of detrimental pathways. CONCLUSIONS: Our findings establish a comprehensive kinase atlas for liver fibrosis, which identifies analogous signaling events conserved among humans and rodents.


Subject(s)
Liver Diseases , Receptor, Insulin , Humans , Mice , Animals , Receptor, Insulin/metabolism , Rodentia , Liver Cirrhosis/pathology , Liver/pathology , Liver Diseases/pathology , Fibrosis , Protein Kinases/metabolism , Collagen/metabolism , Serine/metabolism , Discoidin Domain Receptors/metabolism , Threonine/metabolism
13.
Cells ; 10(2)2021 02 18.
Article in English | MEDLINE | ID: mdl-33670586

ABSTRACT

Bone metastases frequently occur in breast cancer patients and lack appropriate treatment options. Hence, understanding the molecular mechanisms involved in the multistep process of breast cancer bone metastasis and tumor-induced osteolysis is of paramount interest. The serine/threonine kinase AKT plays a crucial role in breast cancer bone metastasis but the effect of individual AKT isoforms remains unclear. Therefore, AKT isoform-specific knockdowns were generated on the bone-seeking MDA-MB-231 BO subline and the effect on proliferation, migration, invasion, and chemotaxis was analyzed by live-cell imaging. Kinome profiling and Western blot analysis of the TGFß/CTGF axis were conducted and metastasis was evaluated by intracardiac inoculation of tumor cells into NOD scid gamma (NSG) mice. MDA-MB-231 BO cells exhibited an elevated AKT3 kinase activity in vitro and responded to combined treatment with AKT- and mTOR-inhibitors. Knockdown of AKT3 significantly increased migration, invasion, and chemotaxis in vitro and metastasis to bone but did not significantly enhance osteolysis. Furthermore, knockdown of AKT3 increased the activity and phosphorylation of pro-metastatic HER2 and DDR1/2 but lowered protein levels of CTGF after TGFß-stimulation, an axis involved in tumor-induced osteolysis. We demonstrated that AKT3 plays a crucial role in bone-seeking breast cancer cells by promoting metastatic potential without facilitating tumor-induced osteolysis.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/metabolism , Connective Tissue Growth Factor/metabolism , Discoidin Domain Receptors/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/metabolism , Transforming Growth Factor beta/metabolism , Animals , Antibodies, Heterophile , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Enzyme Activation , Female , Gene Knockdown Techniques , Humans , Mice , Neoplasm Metastasis , Proto-Oncogene Proteins c-akt/deficiency , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction
14.
Cartilage ; 13(2_suppl): 1285S-1291S, 2021 12.
Article in English | MEDLINE | ID: mdl-31177815

ABSTRACT

One of the most pressing issues in osteoarthritis (OA) research is the development of disease-modifying OA drugs (DMOADs), as currently there are no such drugs available. The paucity of suitable DMOADs is mostly due to the lack of approved ideal therapeutic targets necessary for the development of these drugs. However, based on recent discoveries from our laboratory and other independent laboratories, it is indicated that a cell surface receptor tyrosine kinase for collagen type II, discoidin domain receptor 2 (DDR2), may be an ideal therapeutic target for the development of DMOADs. In this article, we review the current status of research in understanding roles of DDR2 in the development of OA.


Subject(s)
Cartilage, Articular , Discoidin Domain Receptor 2 , Osteoarthritis , Cartilage, Articular/metabolism , Discoidin Domain Receptor 2/metabolism , Discoidin Domain Receptors/metabolism , Humans , Matrix Metalloproteinase 13/metabolism , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Receptors, Mitogen/metabolism
15.
Cancer Sci ; 112(3): 962-969, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33377205

ABSTRACT

Discoidin domain receptors (DDR), including DDR1 and DDR2, are special types of the transmembrane receptor tyrosine kinase superfamily. DDR are activated by binding to the triple-helical collagen and, in turn, DDR can activate signal transduction pathways that regulate cell-collagen interactions involved in multiple physiological and pathological processes such as cell proliferation, migration, apoptosis, and cytokine secretion. Recently, DDR have been found to contribute to various diseases, including cancer. In addition, aberrant expressions of DDR have been reported in various human cancers, which indicates that DDR1 and DDR2 could be new targets for cancer treatment. Considerable effort has been made to design DDR inhibitors and several molecules have shown therapeutic effects in pre-clinical models. In this article, we review the recent literature on the role of DDR in cancer progression, the development status of DDR inhibitors, and the clinical potential of targeting DDR in cancer therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Discoidin Domain Receptors/metabolism , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Carcinogenesis/drug effects , Carcinogenesis/pathology , Cell Line, Tumor , Discoidin Domain Receptors/antagonists & inhibitors , Discoidin Domain Receptors/genetics , Disease Models, Animal , Disease Progression , Drug Screening Assays, Antitumor , Humans , Mice , Mice, Knockout , Molecular Targeted Therapy/methods , Neoplasms/pathology , Protein Kinase Inhibitors/therapeutic use
16.
Blood ; 136(14): 1590-1598, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32746453

ABSTRACT

The discovery of clonal hematopoiesis (CH) in older individuals has changed the way hematologists and stem cell biologists view aging. Somatic mutations accumulate in stem cells over time. While most mutations have no impact, some result in subtle functional differences that ultimately manifest in distinct stem cell behaviors. With a large pool of stem cells and many decades to compete, some of these differences confer advantages under specific contexts. Approximately 20 genes are recurrently found as mutated in CH, indicating they confer some advantage. The impact of these mutations has begun to be analyzed at a molecular level by modeling in cell lines and in mice. Mutations in epigenetic regulators such as DNMT3A and TET2 confer an advantage by enhancing self-renewal of stem and progenitor cells and inhibiting their differentiation. Mutations in other genes involved in the DNA damage response may simply enhance cell survival. Here, we review proposed mechanisms that lead to CH, specifically in the context of stem cell biology, based on our current understanding of the function of some of the CH-associated genes.


Subject(s)
Clonal Hematopoiesis , Hematopoiesis , Stem Cells/cytology , Stem Cells/metabolism , Animals , Biomarkers , Cell Differentiation/genetics , Clonal Evolution/genetics , Clonal Hematopoiesis/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Damage , DNA Methyltransferase 3A , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dioxygenases , Discoidin Domain Receptors/genetics , Discoidin Domain Receptors/metabolism , Gene Expression Regulation, Developmental , Hematopoiesis/genetics , Humans , Mutation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism
17.
J Bone Miner Metab ; 38(5): 670-677, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32415375

ABSTRACT

INTRODUCTION: The periosteum has a bilayered structure that surrounds cortical bone. The outer layer is rich in connective tissue and fibroblasts, while the inner layer in contact with the cortical surface of the bone predominantly consists of osteoblasts and osteoblast progenitors. The identification of cell-specific surface markers of the bilayered structure of the periosteum is important for the purpose of tissue regeneration. MATERIALS AND METHODS: We investigated the expression of the discoidin domain tyrosine kinase receptor DDR2, fibroblast specific protein-1 (FSP-1) and alkaline phosphatase (ALP) in the periosteum of cortical bone by immunohistochemistry. Osteogenic differentiation was compared between DDR2- and FSP-1-expressing cells flow-sorted from the periosteum. RESULTS: We showed that DDR2 predominantly labeled osteogenic cells residing in the inner layer of the periosteum and that Pearson's coefficient of colocalization indicated a significant correlation with the expression of ALP. The mineralization of DDR2-expressing osteogenic cells isolated from the periosteum was significantly induced. In contrast, FSP-1 predominantly labeled the outer layer of periosteal fibroblasts, and Pearson's coefficient of colocalization indicated that FSP-1 was poorly correlated with the expression of DDR2 and ALP. FSP-1-expressing periosteal fibroblasts did not exhibit osteogenic differentiation for the induction of bone mineralization. CONCLUSION: DDR2 is a novel potential cell surface marker for identifying and isolating osteoblasts and osteoblast progenitors within the periosteum that can be used for musculoskeletal regenerative therapies.


Subject(s)
Discoidin Domain Receptors/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Periosteum/cytology , Alkaline Phosphatase/metabolism , Animals , Biomarkers/metabolism , Calcification, Physiologic , Cell Differentiation , Mice, Inbred C57BL , Osteogenesis , S100 Calcium-Binding Protein A4/metabolism
19.
Biochim Biophys Acta Mol Cell Res ; 1866(11): 118522, 2019 11.
Article in English | MEDLINE | ID: mdl-31394114

ABSTRACT

The insulin/insulin-like growth factor system (IIGFs) plays a fundamental role in the regulation of prenatal and postnatal growth, metabolism and homeostasis. As a consequence, dysregulation of this axis is associated with growth disturbance, type 2 diabetes, chronic inflammation and tumor progression. A functional crosstalk between IIGFs and discoidin domain receptors (DDRs) has been recently discovered. DDRs are non-integrin collagen receptors that canonically undergo slow and long-lasting autophosphorylation after binding to fibrillar collagen. While both DDR1 and DDR2 functionally interact with IIGFs, the crosstalk with DDR1 is so far better characterized. Notably, the IIGFs-DDR1 crosstalk presents a feed-forward mechanism, which does not require collagen binding, thus identifying novel non-canonical action of DDR1. Further studies are needed to fully explore the role of this IIGFs-DDRs functional loop as potential target in the treatment of inflammatory and neoplastic disorders.


Subject(s)
Discoidin Domain Receptors/metabolism , Insulin/metabolism , Somatomedins/metabolism , Animals , Diabetes Mellitus, Type 2 , Discoidin Domain Receptor 1/metabolism , Discoidin Domain Receptor 2/metabolism , Fibrosis , Humans , Inflammation , Insulin-Like Growth Factor II/metabolism , Neoplasms/metabolism , Phosphorylation , Protein Binding , Protein Isoforms , Receptor, Insulin , Receptors, Somatomedin , Signal Transduction , Thyroid Neoplasms/metabolism
20.
EMBO Rep ; 20(9): e47250, 2019 09.
Article in English | MEDLINE | ID: mdl-31338941

ABSTRACT

Despite playing physiological roles in specific situations, DNA-RNA hybrids threat genome integrity. To investigate how cells do counteract spontaneous DNA-RNA hybrids, here we screen an siRNA library covering 240 human DNA damage response (DDR) genes and select siRNAs causing DNA-RNA hybrid accumulation and a significant increase in hybrid-dependent DNA breakage. We identify post-replicative repair and DNA damage checkpoint factors, including those of the ATM/CHK2 and ATR/CHK1 pathways. Thus, spontaneous DNA-RNA hybrids are likely a major source of replication stress, but they can also accumulate and menace genome integrity as a consequence of unrepaired DSBs and post-replicative ssDNA gaps in normal cells. We show that DNA-RNA hybrid accumulation correlates with increased DNA damage and chromatin compaction marks. Our results suggest that different mechanisms can lead to DNA-RNA hybrids with distinct consequences for replication and DNA dynamics at each cell cycle stage and support the conclusion that DNA-RNA hybrids are a common source of spontaneous DNA damage that remains unsolved under a deficient DDR.


Subject(s)
DNA Damage/physiology , Cell Cycle/genetics , Cell Cycle/physiology , DNA Damage/genetics , DNA Replication/genetics , DNA Replication/physiology , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Discoidin Domain Receptors/genetics , Discoidin Domain Receptors/metabolism , Flow Cytometry , HeLa Cells , Humans , Models, Biological , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL