Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Bioorg Chem ; 132: 106321, 2023 03.
Article in English | MEDLINE | ID: mdl-36642020

ABSTRACT

Osteoarthritis (OA), a degenerative disease affecting the joint, is characterized by degradation of the joint edge, cartilage injury, and subchondral bone hyperplasia. Treatment of early subchondral bone loss in OA can inhibit subsequent articular degeneration and improve the prognosis of OA. PD0325901, a specific inhibitor of ERK, is widely used in oncology and has potential as a therapeutic agent for osteoarthritis In this study, we investigated the biological function of PD0325901 in bone marrow monocytes/macrophages (BMMs)treated with RANKL and found that it inhibited osteoclast differentiation in vitro in a time- and dose-dependent manner. PD0325901 restrained the expression of osteoclast marker genes, such as c-Fos and NFATc1 induced by RANKL. We tested the biological effects of PD035901 on ATDC5 cells stimulated by IL-1ß and found that it had protective effects on ATDC5 cells. In animal studies, we used a destabilization of the medial meniscus (DMM) model and injected 5 mg/kg or 10 mg/kg of PD0325901 compound into each experimental group of mice. We found that PD0325901 significantly reduced osteochondral pathological changes in post-OA subchondral bone destruction.Finally, we found that PD0325901 down-regulated the pyroptosis level in chondrocytes to rescue cartilage degeneration. Therefore, PD0325901 is expected to be a new generation alternative therapy for OA.


Subject(s)
NF-kappa B , Osteoarthritis , Animals , Mice , NF-kappa B/metabolism , Osteoclasts , Signal Transduction , Inflammation/metabolism , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Cartilage/metabolism , Cartilage/pathology , Chondrocytes
2.
Exp Eye Res ; 218: 109012, 2022 05.
Article in English | MEDLINE | ID: mdl-35245513

ABSTRACT

Aniridia is a panocular condition characterized by impaired eye development and vision, which is mainly due to the haploinsufficiency of the paired-box-6 (PAX6) gene. Like what is seen in aniridia patients, Pax6-deficient mice Pax6Sey-Neu/+ exhibit a varied degree of ocular damage and impaired vision. Our previous studies showed that these phenotypes were partially rescued by PD0325901, a mitogen-activated protein kinase kinase (MEK or MAP2K) inhibitor. In this study, we assessed the long-term efficacy of PD0325901 treatment in retinal health and visual behavior. At about one year after the postnatal treatment with PD0325901, Pax6Sey-Neu/+ mice showed robust improvements in retina size and visual acuity, and the elevated intraocular pressure (IOP) was also alleviated, compared to age-matched mice treated with vehicles only. Moreover, the Pax6Sey-Neu/+ eyes showed disorganized retinal ganglion cell (RGC) axon bundles and retinal layers, which we termed as hotspots. We found that the PD treatment reduced the number and size of hotspots in the Pax6Sey-Neu/+ retinas. Taken together, our results suggest that PD0325901 may serve as an efficacious intervention in protecting retina and visual function in aniridia-afflicted subjects.


Subject(s)
Aniridia , Paired Box Transcription Factors , Animals , Aniridia/genetics , Disease Models, Animal , Eye Proteins/genetics , Haploinsufficiency , Homeodomain Proteins/genetics , Humans , Mice , Mitogen-Activated Protein Kinase Kinases/genetics , PAX6 Transcription Factor/genetics , Paired Box Transcription Factors/genetics , Repressor Proteins/genetics , Retina
3.
Acta Pharm Sin B ; 11(10): 3120-3133, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34729305

ABSTRACT

ERK pathway regulated the programmed death ligand-1 (PD-L1) expression which was linked to the response of programmed death-1 (PD-1)/PD-L1 blockade therapy. So it is deducible that ERK inhibitor could enhance the efficacy of PD-1 inhibitor in cancer immunotherapy. In this study, PD0325901, an oral potent ERK inhibitor, strongly enhanced the efficacy of PD-1 antibody in vitro and in vivo models in non-small cell lung carcinoma (NSCLC) cells. Mechanistically, PD0325901 or shRNA-ERK1/2 significantly downregulated the PD-L1 expression in NSCLC cells and increased the CD3+ T cells infiltration and functions in tumor tissue. There was a positive correlation between the p-ERK1/2 expression and PD-L1 expression in patients with NSCLC. And the patients with low p-ERK1/2 expression were observed a high response rate of PD-1/PD-L1 blockage therapy. Our results demonstrate that PD0325901, an ERK inhibitor, can enhance the efficacy of PD-1 blockage against NSCLC in vitro and in vivo models. And the combination of ERK inhibitor such as PD0325901 and PD-1/PD-L1 blockage is a promising regimen and encouraged to be further confirmed in the treatment of patients with NSCLC.

4.
Acta Pharmaceutica Sinica B ; (6): 3120-3133, 2021.
Article in English | WPRIM (Western Pacific) | ID: wpr-922784

ABSTRACT

ERK pathway regulated the programmed death ligand-1 (PD-L1) expression which was linked to the response of programmed death-1 (PD-1)/PD-L1 blockade therapy. So it is deducible that ERK inhibitor could enhance the efficacy of PD-1 inhibitor in cancer immunotherapy. In this study, PD0325901, an oral potent ERK inhibitor, strongly enhanced the efficacy of PD-1 antibody

5.
Clin Sci (Lond) ; 133(16): 1797-1811, 2019 08 30.
Article in English | MEDLINE | ID: mdl-31387935

ABSTRACT

Background: Aneurysmal subarachnoid haemorrhage (SAH) is a variant of haemorrhagic stroke with a striking 50% mortality rate. In addition to the initial insult, secondary delayed brain injury may occur days after the initial ischemic insult and is associated with vasospasms leading to delayed cerebral ischemia. We have previously shown that the MEK1/2 inhibitor U0126 improves neurological assessment after SAH in rats. Aim: The purpose of the present study was to analyse the impact of a broad selection of high potency MEK1/2 inhibitors in an organ culture model and use the IC50 values obtained from the organ culture to select highly potent inhibitors for pre-clinical in vivo studies. Results: Nine highly potent mitogen activated protein kinase kinase (MEK1/2) inhibitors were screened and the two most potent inhibitors from the organ culture screening, trametinib and PD0325901, were tested in an in vivo experimental rat SAH model with intrathecal injections. Subsequently, the successful inhibitor trametinib was administered intraperitoneally in a second in vivo study. In both regimens, trametinib treatment caused significant reductions in the endothelin-1 induced contractility after SAH, which is believed to be associated with endothelin B receptor up-regulation. Trametinib treated rats showed improved neurological scores, evaluated by the ability to traverse a rotating pole, after induced SAH. Conclusion: The PD0325901 treatment did not improve the neurological score after SAH, nor showed any beneficial therapeutic effect on the contractility, contrasting with the reduction in neurological deficits seen after trametinib treatment. These data show that trametinib might be a potential candidate for treatment of SAH.


Subject(s)
Cerebral Arteries/drug effects , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Subarachnoid Hemorrhage/drug therapy , Animals , Basilar Artery/drug effects , Basilar Artery/metabolism , Benzamides/pharmacology , Benzamides/therapeutic use , Cerebral Arteries/metabolism , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Diphenylamine/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Male , Muscle Contraction/drug effects , Organ Culture Techniques , Pyridones/pharmacology , Pyridones/therapeutic use , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , Rats , Rats, Sprague-Dawley
6.
Am J Cancer Res ; 9(6): 1282-1292, 2019.
Article in English | MEDLINE | ID: mdl-31285959

ABSTRACT

ErbB family members that contain EGFR, HER2, HER3 and HER4 play important roles in many cancer types, including head and neck; however, inhibition of these receptors by small molecule kinase inhibitors showed limited results due to compensatory up-regulation of some key survival signaling pathways. Here, we explore the effectiveness of Afatinib, an irreversible inhibitor of EGFR, HER2, and HER4, in combination with the MEK inhibitor PD0325901 to inhibit cisplatin-resistant head and neck squamous cell carcinoma (HNSCC). We treated two cisplatin-resistant HNSCC cell lines, UMSCC74B and O28, with Afatinib, PD0325901, or a combination, and measured signaling pathways, cell proliferation, and survival. We found that Afatinib blocked Akt/mTOR activity and phosphorylation of EGFR, HER2 and HER3, but up-regulated MEK/ERK signaling. Interestingly, MEK inhibitor PD0325901 blocked ERK phosphorylation, but elevated phosphorylation of Akt and mTOR pathways. Similarly, Afatinib and PD0325901 inhibited all these pathways and synergistically suppressed cell proliferation and survival. Our data demonstrate that Afatinib in combination with MEK inhibitors could provide a potential novel therapy for cisplatin-resistant head and neck squamous cell cancer.

7.
Endocr Relat Cancer ; 26(9): 739-750, 2019 09.
Article in English | MEDLINE | ID: mdl-31272080

ABSTRACT

Anaplastic thyroid cancer (ATC) is an aggressive malignancy with limited treatment options. We explored novel treatment modalities by targeting epigenetic modifications using inhibitors of BET (e.g. BRD4) activity. We evaluated the efficacy in the treatment of ATC of a novel BET inhibitor, PLX51107 (PLX), currently in clinical trials for other solid tumors and hematologic malignancies, alone or combined with a MEK inhibitor, PD0325901(PD). To elucidate the effects of these inhibitors on growth of ATC, we treated ATC cells derived from patient tumors (THJ-11T and THJ-16T cells) and mouse xenograft tumors with inhibitors. We found PLX and PD inhibitors singly inhibited proliferation of both human ATC cells lines, but together exhibited stronger inhibition of proliferation. In mouse xenografts, the combination treatment almost totally blocked growth in xenograft tumors derived from both ATC cells. PD effectively attenuated MEK-ERK signaling, which was further enhanced by PLX in the combined treatment in cultured cells and tumors. Importantly, the combination of PLX and PD acted synergistically to suppress MYC transcription to increase p27 in decreasing tumor cell proliferation. PLX and PD cooperated to upregulate pro-apoptotic proteins to promote apoptosis. These two inhibitors converged to reduce the binding of BRD4 to the MYC promoter to suppress the MYC expression. These findings indicate that combined treatment of BET and MEK-ERK inhibitors was more effective to treat ATC than single targeted treatment. Synergistic suppression of MYC transcription via collaborative actions on chromatin modifications suggested that targeting epigenetic modifications could provide novel treatment opportunities for ATC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzamides/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Diphenylamine/analogs & derivatives , MAP Kinase Kinase 1/antagonists & inhibitors , Oxazoles/pharmacology , Pyridines/pharmacology , Pyrroles/pharmacology , Thyroid Carcinoma, Anaplastic/drug therapy , Thyroid Neoplasms/drug therapy , Transcription Factors/antagonists & inhibitors , Animals , Apoptosis/drug effects , Benzamides/administration & dosage , Cell Proliferation/drug effects , Diphenylamine/administration & dosage , Diphenylamine/pharmacology , Drug Synergism , Female , Humans , Mice , Mice, Nude , Oxazoles/administration & dosage , Pyridines/administration & dosage , Pyrroles/administration & dosage , Thyroid Carcinoma, Anaplastic/enzymology , Thyroid Carcinoma, Anaplastic/metabolism , Thyroid Carcinoma, Anaplastic/pathology , Thyroid Neoplasms/enzymology , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
BMC Dev Biol ; 19(1): 13, 2019 07 04.
Article in English | MEDLINE | ID: mdl-31272387

ABSTRACT

BACKGROUND: The segregation of the hypoblast and the emergence of the pluripotent epiblast mark the final stages of blastocyst formation in mammalian embryos. In bovine embryos the formation of the hypoblast has been partially studied, and evidence shows that MEK signalling plays a limited role in the segregation of this lineage. Here we explored the role of different signalling pathways during lineage segregation in the bovine embryo using immunofluorescence analysis of NANOG and SOX17 as readouts of epiblast and hypoblast, respectively. RESULTS: We show that SOX17 starts to be expressed in 16-32-cell stage embryos, whereas NANOG is first detected from 8-cell stage. SOX17 is first co-expressed with NANOG, but these markers become mutually exclusive by the late blastocyst stage. By assessing the expression kinetics of NANOG/SOX17 we show that inhibition of MEK signalling can eliminate SOX17 expression in bovine blastocysts, without altering NANOG expression. Modulation of WNT, PKC and LIF did not affect NANOG expression in the epiblast when used in combination with the ERK inhibitor. CONCLUSIONS: This study shows that SOX17 can be used as a reliable early marker of hypoblast in the bovine, and based on its expression profile we show that the hypoblast segregates in day 7 blastocysts. Furthermore, SOX17 expression is abolished using 1 µM of PD0325901, without affecting the NANOG population in the epiblast. Modulation of WNT, PKC and LIF are not sufficient to support enhanced NANOG expression in the epiblast when combined with ERK inhibitor, indicating that additional signalling pathways should be examined to determine their potential roles in epiblast expansion.


Subject(s)
Blastocyst/cytology , Embryo, Mammalian/embryology , Germ Layers/embryology , Nanog Homeobox Protein/metabolism , SOXF Transcription Factors/metabolism , Animals , Benzamides/pharmacology , Cattle , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Germ Layers/cytology , Leukemia Inhibitory Factor/biosynthesis , Nanog Homeobox Protein/genetics , Protein Kinase C/biosynthesis , SOXF Transcription Factors/genetics , Signal Transduction/physiology , Wnt1 Protein/biosynthesis
9.
Pigment Cell Melanoma Res ; 32(6): 829-841, 2019 11.
Article in English | MEDLINE | ID: mdl-31251472

ABSTRACT

RAS is frequently mutated in various tumors and known to be difficult to target. NRASQ61K/R are the second most frequent mutations found in human skin melanoma after BRAFV600E . Aside from surgery, various approaches, including targeted therapies, immunotherapies, and combination therapies, are used to treat patients carrying NRAS mutations, but they are inefficient. Here, we established mouse NRASQ61K melanoma cell lines and genetically derived isografts (GDIs) from Tyr::NRASQ61K mouse melanoma that can be used in vitro and in vivo in an immune-competent environment (C57BL/6) to test and discover novel therapies. We characterized these cell lines at the cellular, molecular, and oncogenic levels and show that NRASQ61K melanoma is highly sensitive to the combination of Mek and Akt inhibitors. This preclinical model shows much potential for the screening of novel therapeutic strategies for patients harboring NRAS mutations that have limited therapeutic options and resulted in poor prognoses.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Melanoma/pathology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Monomeric GTP-Binding Proteins/genetics , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzimidazoles/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Shape/drug effects , Heterocyclic Compounds, 3-Ring/pharmacology , Melanocytes/drug effects , Melanocytes/pathology , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase Kinases/metabolism , Models, Biological , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
10.
BMC Res Notes ; 11(1): 520, 2018 Jul 28.
Article in English | MEDLINE | ID: mdl-30055648

ABSTRACT

OBJECTIVE: Malignant peripheral nerve sheath tumors (MPNST) are aggressive sarcomas characterized by high recurrence rates and early metastases. These tumors arise more frequently within neurofibromatosis type 1 (NF1) and present with resistance during standard chemotherapy leading to increased mortality and morbidity in those patients. In vitro all-trans retinoic acid (ATRA) and MEK inhibitors (MEKi) were shown to inhibit tumor proliferation, especially when applied in combination. Therefore, we established a nude mouse model to investigate if treatment of xenografts derived from NF1 associated S462 and T265 MPNST cells respond to ATRA and the MEKi PD0325901. RESULTS: We demonstrated that human NF1 associated MPNST derived from S462 but not T265 cells form solid subcutaneous tumors in Foxn1 nude mice but not in Balb/c, SHO or Shorn mice. We verified a characteristic staining pattern of human MPNST xenografts by immunohistochemistry. Therapeutic effects of ATRA and/or MEKi PD0325901 on growth of S462 MPNST xenografts in Foxn1 nude mice were not demonstrated in vitro, as we did not observe significant suppression of MPNST growth compared with placebo treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Diphenylamine/analogs & derivatives , Nerve Sheath Neoplasms/drug therapy , Sarcoma/drug therapy , Animals , Diphenylamine/pharmacology , Heterografts , Humans , Mice , Mice, Nude , Neurilemmoma
11.
Pharmacol Rep ; 70(4): 777-783, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29940508

ABSTRACT

BACKGROUND: Synaptic Ras-GTPase-activating protein 1 (SYNGAP1) is an abundant brain-specific protein localized at the postsynaptic density of mammalian excitatory synapses. SYNGAP1 functions as a crucial regulator of downstream intracellular signaling triggered by N-methyl-d-aspartate receptor activation. One of the most important signaling pathways regulated by SYNGAP1 is the Ras-Raf-MEK-ERK pathway. SYNGAP1 deficiency is associated with hyperphosphorylation of MEK and ERK kinases and with altered synaptic function in Syngap1+/- mice. Loss-of-function mutations in the SYNGAP1 gene have been documented in many human cognitive and neurological disorders. However, there are currently no approaches that reverse the phenotypes of SYNGAP1 deficiency. METHODS: Using electrophysiological recordings of field responses in hippocampal slices, we examined if disturbances of synaptic physiology in the hippocampus of 7-8-month old Syngap1+/- mice were sensitive to the effect of the MEK inhibitor PD-0325901 given orally for 6days. RESULTS: We found that in hippocampal slices from vehicle-treated Syngap1+/- mice, basal synaptic responses were higher and their long-term potentiation (LTP) was lower than in slices from wild-type littermates. Chronic administration of PD-0325901 normalized basal synaptic responses, but did not reverse LTP deficit. CONCLUSIONS: The differential sensitivity of basal synaptic transmission and LTP to MEK inhibition indicates that the effects of SYNGAP1 deficiency on these synaptic parameters are mediated by distinct pathways. Our findings also suggest that at least some physiological phenotypes of the germline Syngap1 mutation can be ameliorated by pharmacological treatment of adult animals.


Subject(s)
Benzamides/pharmacology , Diphenylamine/analogs & derivatives , Hippocampus/physiopathology , Membrane Potentials/drug effects , ras GTPase-Activating Proteins/deficiency , Animals , Diphenylamine/pharmacology , Female , Long-Term Potentiation/drug effects , Male , Mice , Mutation , ras GTPase-Activating Proteins/genetics
12.
J Ocul Pharmacol Ther ; 34(6): 477-485, 2018.
Article in English | MEDLINE | ID: mdl-29708810

ABSTRACT

PURPOSE: ST-162 and ST-168 are small-molecule bifunctional inhibitors of MEK and PI3K signaling pathways that are being developed as novel antitumor agents. Previous small-molecule and biologic MEK inhibitors demonstrated ocular toxicity events that were dose limiting in clinical studies. We evaluated in vitro and in vivo ocular toxicity profiles of ST-162 and ST-168. METHODS: Photoreceptor cell line 661W and adult retinal pigment epithelium cell line ARPE-19 were treated with increasing concentrations of bifunctional inhibitors. Western blots, cell viability, and caspase activity assays were performed to evaluate MEK and PI3K inhibition and dose-dependent in vitro toxicity, and compared with monotherapy. In vivo toxicity profile was assessed by intravitreal injection of ST-162 and ST-168 in Dutch-Belted rabbits, followed by ocular examination and histological analysis of enucleated eyes. RESULTS: Retinal cell lines treated with ST-162 or ST-168 exhibited dose-dependent inhibition of MEK and PI3K signaling. Compared with inhibition by monotherapies and their combinations, bifunctional inhibitors demonstrated reduced cell death and caspase activity. In vivo, both bifunctional inhibitors exhibited a more favorable toxicity profile when compared with MEK inhibitor PD0325901. CONCLUSIONS: Novel MEK and PI3K bifunctional inhibitors ST-162 and ST-168 demonstrate favorable in vitro and in vivo ocular toxicity profiles, supporting their further development as potential therapeutic agents targeting multiple aggressive tumors.


Subject(s)
Eye/drug effects , MAP Kinase Kinase Kinases/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/adverse effects , Visual Acuity/drug effects , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Humans , Intravitreal Injections , MAP Kinase Kinase Kinases/metabolism , Male , Mice , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Rabbits
13.
Acta Histochem ; 120(2): 142-150, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29397960

ABSTRACT

Chondrosarcoma is a malignant bone tumor that produces cartilaginous neoplastic tissue. Owing to the absence of an effective adjuvant therapy, high-grade chondrosarcoma has a poor prognosis. Therefore, it is important to develop an effective adjuvant therapy to prevent the recurrence and metastasis. Mammalian target of rapamycin (mTOR), a central regulator of cell growth, metabolism, proliferation, and survival, is considered an important target for anticancer drug development. The mitogen activated protein kinase (MAPK) pathway is another highly implicated cellular pathway in cancer and is thought to have compensatory effects in response to the inhibition of the phosphatidylinositol-3-kinase (PI3K)/Akt/mTOR signaling pathway. We investigated the mechanism of anti-proliferative effect of the mTOR inhibitor rapamycin and MAPK/ERK (MEK) inhibitor PD 0325901, and the combined effect of rapamycin and PD 0325901 on human chondrosarcoma cell line (OUMS-27). Combination therapy with rapamycin and PD 0325901 showed a stronger anti-proliferative effect on OUMS-27 cells than rapamycin monotherapy. We confirmed that the dual inhibition of the PI3K/Akt/mTOR and RAF/MEK/ERK signaling pathways had synergistic anti-proliferative effects in OUMS-27. Our results suggest that combination therapy of mTOR and MEK inhibitor could be an effective therapeutic approach against chondrosarcoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Synergism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/classification , Blotting, Western , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Flow Cytometry , Humans , MAP Kinase Signaling System/drug effects
14.
Stem Cell Res Ther ; 9(1): 2, 2018 01 05.
Article in English | MEDLINE | ID: mdl-29304842

ABSTRACT

BACKGROUND: Pluripotent stem cells hold great promise for regenerative medicine. However, before clinical application, reproducible protocols for pluripotent stem cell differentiation should be established. Extracellular signal-regulated protein kinase (ERK) signaling plays a central role for the self-renewal of epiblast stem cells (EpiSCs), but its role for subsequent germ layer differentiation is still ambiguous. We proposed that ERK could modulate differentiation of the epiblast. METHODS: PD0325901 was used to inhibit ERK activation during the differentiation of embryonic stem cells and EpiSCs. Immunofluorescence, western blot analysis, real-time PCR and flow cytometry were used to detect germ layer markers and pathway activation. RESULTS: We demonstrate that the ERK phosphorylation level is lower in neuroectoderm of mouse E7.5 embryos than that in the primitive streak. ERK inhibition results in neural lineage commitment of epiblast. Mechanistically, PD0325901 abrogates the expression of primitive streak markers by ß-catenin retention in the cytoplasm, and inhibits the expression of OCT4 and NANOG during EpiSC differentiation. Thus, EpiSCs differentiate into neuroectodermal lineage efficiently under PD0325901 treatment. These results suggest that neuroectoderm differentiation does not require extrinsic signals, supporting the default differentiation of neural lineage. CONCLUSIONS: We report that a single ERK inhibitor, PD0325901, can specify epiblasts and EpiSCs into neural-like cells, providing an efficient strategy for neural differentiation.


Subject(s)
Embryonic Stem Cells/cytology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Germ Layers/cytology , Neural Plate/cytology , Neurogenesis/physiology , Primitive Streak/cytology , Animals , Benzamides/pharmacology , Cells, Cultured , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nanog Homeobox Protein/biosynthesis , Neural Plate/metabolism , Octamer Transcription Factor-3/biosynthesis , Octamer Transcription Factor-3/genetics , Phosphorylation , Primitive Streak/metabolism , beta Catenin/metabolism
15.
Biochem Biophys Res Commun ; 483(1): 325-331, 2017 01 29.
Article in English | MEDLINE | ID: mdl-28025142

ABSTRACT

The identification of small molecular inhibitors, which were reported to promote the derivation of mouse and human embryonic stem cells (ESCs), provides a potential strategy for the derivation of domesticated ungulate ESCs. In present study, goat inner cell mass (ICM) derived cells in the double inhibition (2i) condition, in which, mitogen-activated protein kinase kinase (MAP2K) and glycogen synthase kinase 3 (GSK3) were inhibited by PD0325901 and BIO respectively, were characterized. The results showed that goat ICM derived cells in 2i medium adding leukaemia inhibitor factor (LIF) possessed a mouse ES-like morphology. But these cells had much compromised proliferation capacity, resulting in difficulty in expansion. In 2i alone medium, goat ICM derived cells possessed primate ES-like morphology. These cells expressed pluripotent markers and could differentiate into derivatives of three germ layers in vitro. However, these cells could not be proliferated in long-term (persisted for 15 passages) because of spontaneously neural differentiation. Additionally, goat ICM derived cells could be inducing differentiated into neural lineage in vitro. Although goat ESCs could not be established in PD0325901 and BIO alone medium, this derivation condition provides a useful research system to find signaling molecular those regulate early embryonic development and pluripotency in goat.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Neurons/metabolism , Pluripotent Stem Cells/cytology , Animals , Benzamides/chemistry , Blastocyst/metabolism , Cell Lineage , Cell Proliferation , Coculture Techniques , Diphenylamine/analogs & derivatives , Diphenylamine/chemistry , Fibroblasts/metabolism , Germ Layers , Glycogen Synthase Kinase 3/metabolism , Goats , Leukemia Inhibitory Factor/metabolism , Mice , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
16.
Cell Signal ; 29: 138-149, 2017 01.
Article in English | MEDLINE | ID: mdl-27777072

ABSTRACT

Mutations in KIT or PDGFRA are responsible for >85% of gastrointestinal stromal tumors. The introduction of imatinib in the GIST therapy scheme revolutionized the patient outcome. Unfortunately, the therapy allows the disease stabilization instead of curation. Furthermore the resistance to the inhibitor arises in most cases within two first years of therapy. A thorough investigation of the signalling pathways activated by the major PDGFRA and KIT mutants encountered in the GIST landscape allowed to identify striking differences between the two receptor tyrosine kinases. PDGFRA mutants were not responsive to their ligand, PDGFAA, and displayed a high constitutive kinase activity. In contrast, all KIT mutants retained, in addition to their constitutive activation, the ability to be stimulated by their ligand. Kit mutants displayed a lower intrinsic kinase activity relative to PDGFRA mutants, while the KIT Exon 11 deletion mutant exhibited the highest intrinsic kinase activity among KIT mutants. At the transcriptomic level, the MAPK pathway was established as the most prominent activated pathway, which is commonly up-regulated by all PDGFRA and KIT mutants. Inhibition of this pathway, using the MEK inhibitor PD0325901, reduced the proliferation of GIST primary cells at nanomolar concentrations. Altogether, our data demonstrate the high value of MEK inhibitors for combination therapy in GIST treatment and more importantly the interest of evaluating the SCF expression profile in GIST patients presenting KIT mutations.


Subject(s)
Gastrointestinal Stromal Tumors/metabolism , Gastrointestinal Stromal Tumors/pathology , Signal Transduction , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Gastrointestinal Stromal Tumors/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Humans , Ligands , MAP Kinase Signaling System/drug effects , Molecular Dynamics Simulation , Mutation/genetics , Protein Kinase Inhibitors/pharmacology , Protein Transport , Proto-Oncogene Proteins c-kit/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects , Up-Regulation/genetics
17.
Data Brief ; 9: 828-838, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27872880

ABSTRACT

The present data are related to the article entitled "Insights into ligand stimulation effects on gastro-intestinal stromal tumors signaling" (C. Bahlawane, M. Schmitz, E. Letellier, K. Arumugam, N. Nicot, P.V. Nazarov, S. Haan, 2016) [1]. Constitutive and ligand-derived signaling pathways mediated by KIT and PDGFRA mutated proteins found in gastrointestinal stromal tumors (GIST) were compared. Expression of mutant proteins was induced by doxycycline in an isogenic background (Hek293 cells). Kit was identified by FACS at the cell surface and found to be quickly degraded or internalized upon SCF stimulation for both Kit Wild type and Kit mutant counterparts. Investigation of the main activated pathways in GIST unraveled a new feature specific for oncogenic KIT mutants, namely their ability to be further activated by Kit ligand, the stem cell factor (scf). We were also able to identify the MAPK pathway as the most prominent target for a common inhibition of PDGFRA and KIT oncogenic signaling. Western blotting and micro-array analysis were applied to analyze the capacities of the mutant to induce an effective STATs response. Among all Kit mutants, only Kit Ex11 deletion mutant was able to elicit an effective STATs response whereas all PDGFRA were able to do so.

18.
Oncotarget ; 7(26): 39730-39739, 2016 Jun 28.
Article in English | MEDLINE | ID: mdl-27213595

ABSTRACT

A rationally selected combination of small-molecule chemicals can affect cell plasticity and fate, suggesting an open chemistry way to manipulate cells to achieve a specific goal. Here we for the first time demonstrate that a combination of vitamin C (Vc) and PD0325901 can achieve about 90% erasure of 5-methylcytosine (5mC) within 5 days (decreasing from 3.2 to ~ 0.3 5mC per 100 C) in mouse embryonic stem cells (ESCs). The hypomethylated level is comparable to that of gonadal primordial germ cells (PGCs), whose pluripotency is closely associated with the global DNA hypomethylation. In contrast, Vc or PD0325901 alone only induces a moderately reduced level of global DNA methylation. Our mechanistic study suggested that PD0325901 elevated expression of Prdm14, which repressed de novo methyltransferase Dnmt3b and its cofactor Dnmt3l at levels of protein, via the mode to eliminate 5mC from de novo synthesis. By further addition of Vc, the oxidation of 5mC as catalyzed by Tet1/Tet2 dioxygenases was significantly increased as manifested by the elevated level of 5-hydroxymethylcytosine. However, by the depletion of Tet1/Tet2, Vc failed to enhance PD0325901-stimulated hypomethylation of ESCs' genomic DNA. Furthermore, mouse ESCs in Vc/PD0325901-supplemented medium show great morphology and pluripotency. Therefore, we demonstrate a novel and synergistic chemical approach for promoting hypomethylation and sustaining pluripotency of ESCs.


Subject(s)
Ascorbic Acid/pharmacology , Benzamides/pharmacology , DNA Methylation , Diphenylamine/analogs & derivatives , MAP Kinase Kinase Kinases/antagonists & inhibitors , Mouse Embryonic Stem Cells/drug effects , 5-Methylcytosine/chemistry , Animals , Catalysis , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA-Binding Proteins , Diphenylamine/pharmacology , Drug Synergism , Embryonic Stem Cells/cytology , Gene Silencing , Germ Cells/cytology , Mice , Oxygen/chemistry , RNA-Binding Proteins , Transcription Factors/metabolism , DNA Methyltransferase 3B
19.
Int J Pharm ; 505(1-2): 329-40, 2016 May 30.
Article in English | MEDLINE | ID: mdl-27085642

ABSTRACT

Glioblastoma (GBM) is the most aggressive tumor type in the central nervous system. Both tumor-targeting drug delivery and combination therapy of multiple therapeutic agents with distinct mechanisms are important for GBM treatment. We combined these two strategies and developed a new platform of peptide-drug conjugate (RGD-PEG-Suc-PD0325901, W22) for tumor-targeting delivery using a combination of PD0325901 (a MEK1/2 inhibitor) and RGD peptide. In the present study, the combination of PD0325901 and RGD peptide strongly inhibited U87MG model in vitro and in vivo. This inhibition contributed to synergistic suppression of cell proliferation by blocking ERK pathway activity and cell migration. Modified by conjugation strategy, their conjugate W22 enhanced PD0325901 delivery to GBM cells by receptor mediated cellular internalization. W22 showed great superiority in targeting to U87MG xenografted tumors and strong anti-tumor efficacy based on ERK pathway inhibition and tumor-targeted delivery in vitro and in vivo. Moreover, W22 was stable in serum and able to release PD0325901 in the enzymatic environment. These data indicated that the RGD-PEG-Suc-PD0325901 conjugate provided a strategy for effective delivery of PD0325901 and RGD peptide into the GBM cells and inhibition of tumor growth in a synergistic manner.


Subject(s)
Antineoplastic Agents/administration & dosage , Benzamides/administration & dosage , Diphenylamine/analogs & derivatives , Drug Delivery Systems , Glioblastoma/drug therapy , Animals , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Cell Proliferation/drug effects , Central Nervous System Neoplasms/drug therapy , Central Nervous System Neoplasms/pathology , Diphenylamine/administration & dosage , Diphenylamine/pharmacology , Drug Liberation , Drug Synergism , Female , Glioblastoma/pathology , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Mice , Mice, Inbred BALB C , Oligopeptides/chemistry , Xenograft Model Antitumor Assays
20.
Oncotarget ; 6(30): 29991-30005, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26358373

ABSTRACT

Oncogenesis in non-small cell lung cancer (NSCLC) is regulated by a complex signal transduction network. Single-agent targeted therapy fails frequently due to treatment insensitivity and acquired resistance. In this study, we demonstrate that co-inhibition of the MAPK and SRC pathways using a PD0325901 and Saracatinib kinase inhibitor combination can abrogate tumor growth in NSCLC. PD0325901/Saracatinib at 0.25:1 combination was screened against a panel of 28 NSCLC cell lines and 68% of cell lines were found to be sensitive (IC50 < 2 µM) to this combination. In Snail1 positive NSCLC lines, the drug combination complementarily enhanced mesenchymal-epithelial transition (MET), increasing both E-cadherin and Plakoglobin expression, and reducing Snail1, FAK and PXN expression. In addition, the drug combination abrogated cell migration and matrigel invasion. The co-inhibition of MAPK and SRC induced strong G1/G0 cell cycle arrest in the NSCLC lines, inhibited anchorage independent growth and delayed tumor growth in H460 and H358 mouse xenografts. These data provide rationale for further investigating the combination of MAPK and SRC pathway inhibitors in advanced stage NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Lung Neoplasms/drug therapy , MAP Kinase Kinase 1/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , src-Family Kinases/antagonists & inhibitors , Animals , Benzamides/pharmacology , Benzodioxoles/pharmacology , Cadherins/metabolism , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Adhesion/drug effects , Cell Cycle/drug effects , Cell Movement/drug effects , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Female , Humans , Immunoblotting , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , MAP Kinase Kinase 1/metabolism , Mice, Inbred BALB C , Mice, Nude , Microscopy, Confocal , Quinazolines/pharmacology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...