Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 308
Filter
1.
J Invest Dermatol ; 144(7): 1522-1533.e10, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38237731

ABSTRACT

Recessive dystrophic epidermolysis bullosa (RDEB) is a rare skin fragility disorder caused by mutations in COL7A1. RDEB is hallmarked by trauma-induced unremitting blistering, chronic wounds with inflammation, and progressive fibrosis, leading to severe disease complications. There is currently no cure for RDEB-associated fibrosis. Our previous studies and increasing evidence highlighted the profibrotic role of NOTCH pathway in different skin disorders, including RDEB. In this study, we further investigated the role of NOTCH signaling in RDEB pathogenesis and explored the effects of its inhibition by γ-secretase inhibitors DAPT and PF-03084014 (nirogacestat). Our analyses demonstrated that JAG1 and cleaved NOTCH1 are upregulated in primary RDEB fibroblasts (ie, RDEB-derived fibroblasts) compared with controls, and their protein levels are further increased by TGF-ß1 stimulation. Functional assays unveiled the involvement of JAG1/NOTCH1 axis in RDEB fibrosis and demonstrated that its blockade counteracts a variety of fibrotic traits. In particular, RDEB-derived fibroblasts treated with PF-03084014 showed (i) a significant reduction of contractility, (ii) a diminished secretion of TGF-ß1 and collagens, and (iii) the downregulation of several fibrotic proteins. Although less marked than PF-03084014-treated cells, RDEB-derived fibroblasts exhibited a reduction of fibrotic traits also upon DAPT treatment. This study provides potential therapeutic strategies to antagonize RDEB fibrosis onset and progression.


Subject(s)
Amyloid Precursor Protein Secretases , Epidermolysis Bullosa Dystrophica , Fibroblasts , Fibrosis , Jagged-1 Protein , Signal Transduction , Humans , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Epidermolysis Bullosa Dystrophica/drug therapy , Epidermolysis Bullosa Dystrophica/pathology , Epidermolysis Bullosa Dystrophica/genetics , Signal Transduction/drug effects , Fibroblasts/metabolism , Fibroblasts/drug effects , Jagged-1 Protein/metabolism , Jagged-1 Protein/genetics , Down-Regulation/drug effects , Receptor, Notch1/metabolism , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Dipeptides/pharmacology , Collagen Type VII/genetics , Collagen Type VII/metabolism , Cells, Cultured , Skin/pathology , Skin/drug effects , Skin/metabolism , Male , Transforming Growth Factor beta1/metabolism , Female , Diamines , Tetrahydronaphthalenes , Thiazoles , Valine/analogs & derivatives
2.
Angiogenesis ; 26(2): 249-263, 2023 05.
Article in English | MEDLINE | ID: mdl-36376768

ABSTRACT

The Notch signaling pathway is an important therapeutic target for the treatment of inflammatory diseases and cancer. We previously created ligand-specific inhibitors of Notch signaling comprised of Fc fusions to specific EGF-like repeats of the Notch1 extracellular domain, called Notch decoys, which bound ligands, blocked Notch signaling, and showed anti-tumor activity with low toxicity. However, the study of their function depended on virally mediated expression, which precluded dosage control and limited clinical applicability. We have refined the decoy design to create peptibody-based Notch inhibitors comprising the core binding domains, EGF-like repeats 10-14, of either Notch1 or Notch4. These Notch peptibodies showed high secretion properties and production yields that were improved by nearly 100-fold compared to previous Notch decoys. Using surface plasmon resonance spectroscopy coupled with co-immunoprecipitation assays, we observed that Notch1 and Notch4 peptibodies demonstrate strong but distinct binding properties to Notch ligands DLL4 and JAG1. Both Notch1 and Notch4 peptibodies interfere with Notch signaling in endothelial cells and reduce expression of canonical Notch targets after treatment. While prior DLL4 inhibitors cause hyper-sprouting, the Notch1 peptibody reduced angiogenesis in a 3-dimensional in vitro sprouting assay. Administration of Notch1 peptibodies to neonate mice resulted in reduced radial outgrowth of retinal vasculature, confirming anti-angiogenic properties. We conclude that purified Notch peptibodies comprising EGF-like repeats 10-14 bind to both DLL4 and JAG1 ligands and exhibit anti-angiogenic properties. Based on their secretion profile, unique Notch inhibitory activities, and anti-angiogenic properties, Notch peptibodies present new opportunities for therapeutic Notch inhibition.


Subject(s)
Angiogenesis Inhibitors , Endothelial Cells , Receptor, Notch1 , Receptor, Notch4 , Animals , Mice , Angiogenesis Inhibitors/genetics , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Epidermal Growth Factor/metabolism , Immunoprecipitation , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Ligands , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Receptor, Notch4/genetics , Receptor, Notch4/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinal Vessels/drug effects , Surface Plasmon Resonance
3.
BMC Cancer ; 22(1): 558, 2022 May 18.
Article in English | MEDLINE | ID: mdl-35585515

ABSTRACT

BACKGROUND: Laminins are high-molecular weight (400 ~ 900 kDa) proteins in extracellular matrix, which serve as major component of the basal lamina, and play a crucial role in promoting tumor cell migration. This study aimed at characterizing the role of laminin in promoting cancer development, and elucidating the mechanism of tumor progression driven by laminin-Notch signaling in bladder cancer. METHODS: 2D collagen/laminin culture system was established and CCK-8/transwell assay was conducted to evaluate the proliferation/migration ability of Biu-87 and MB49 cells cultured on 2D gels. Activation of integrins-Notch1 signaling was determined by western blotting. Orthotopic bladder cancer mice model was established to assess the therapeutic effects of Notch inhibitor. RESULTS: Our study demonstrated that extracellular laminin can trigger tumor cell proliferation/migration through integrin α6ß4/Notch1 signaling in bladder cancer. Inhibition of Telomere repeat-binding factor 3 (TRB3)/Jagged Canonical Notch Ligand 1 (JAG1) signaling suppressed Notch signals activation induced by laminin-integrin axis. In MB49 orthotopic bladder cancer mice model, Notch inhibitor SAHM1 efficiently improved tumor suppressive effects of chemotherapy and prolonged survival time of tumor-bearing mice. CONCLUSION: In conclusion, we show that, in bladder cancer, extracellular laminin induced the activation of Notch pathway through integrin α6ß4/TRB3/JAG3, and disclosed a novel role of laminin in bladder cancer cells proliferation or migration.


Subject(s)
Integrin alpha6beta4 , Laminin , Urinary Bladder Neoplasms , Animals , Cell Movement , Extracellular Matrix/metabolism , Humans , Integrin alpha6beta4/metabolism , Laminin/metabolism , Mice , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/metabolism , Signal Transduction , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
4.
Microvasc Res ; 140: 104308, 2022 03.
Article in English | MEDLINE | ID: mdl-34995552

ABSTRACT

Intrauterine growth restriction (IUGR) is associated with increased perinatal mortality and morbidity, and plays an important role in the development of adult cardiovascular diseases. This study brings forward a hypothesis that Human umbilical vein endothelial cells (HUVECs) from IUGR newborns present dysfunctions and varying changes of signaling pathways as compared to the Control group. Similar pathways may also be present in pulmonary or systemic vasculatures. HUVECs were derived from newborns. There were three groups according to the different fetal origins: normal newborns (Control), IUGR from poor maternal nutrition (IUGR1), and pregnancy-induced hypertension (IUGR2). We found that IUGR-derived HUVECs showed a proliferative phenotype compared to those from normal subjects. Interestingly, two types IUGR could cause varying degrees of cellular dysfunction. Meanwhile, the Notch1 signaling pathway showed enhanced activation in the two IUGR-induced HUVECs, with subsequent activation of Akt or extracellular signal regulated protein kinases1/2 (ERK1/2). Pharmacological inhibition or gene silencing of Notch1 impeded the proliferative phenotype of IUGR-induced HUVECs and reduced the activation of ERK1/2 and AKT. In summary, elevated Notch1 levels might play a crucial role in IUGR-induced HUVECs disorders through the activation of ERK1/2 and AKT. These pathways could be potential therapeutic targets for prevention of the progression of IUGR associated diseases later in life.


Subject(s)
Fetal Growth Retardation/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Pathologic , Receptor, Notch1/metabolism , Adult , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle Proteins/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Diamines/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fetal Growth Retardation/pathology , Gamma Secretase Inhibitors and Modulators/pharmacology , Gene Silencing , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/pathology , Humans , Infant, Newborn , Phenotype , Phosphorylation , Pregnancy , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Signal Transduction , Thiazoles/pharmacology
5.
Br J Pharmacol ; 179(8): 1716-1731, 2022 04.
Article in English | MEDLINE | ID: mdl-34796471

ABSTRACT

BACKGROUND AND PURPOSE: The Dll4-Notch1 signalling pathway plays an important role in sprouting angiogenesis, vascular remodelling and arterial or venous specificity. Genetic or pharmacological inhibition of Dll4-Notch1 signalling leads to excessive sprouting angiogenesis. However, transcriptional inhibitors of Dll4-Notch1 signalling have not been described. EXPERIMENTAL APPROACH: We designed a new peptide targeting Notch signalling, referred to as TAT-ANK, and assessed its effects on angiogenesis. In vitro, tube formation and fibrin gel bead assay were carried out, using human umbilical vein endothelial cells (HUVECs). In vivo, Matrigel plug angiogenesis assay, a developmental retinal model and tumour models in mice were used. The mechanisms underlying TAT-ANK activity were investigated by immunochemistry, western blotting, immunoprecipitation, RT-qPCR and luciferase reporter assays. KEY RESULTS: The amino acid residues 179-191 in the G-protein-coupled receptor-kinase-interacting protein-1 (GIT1-ankyrin domain) are crucial for GIT1 binding to the Notch transcription repressor, RBP-J. We designed the peptide TAT-ANK, based on residues 179-191 in GIT1. TAT-ANK significantly inhibited Dll4 expression and Notch 1 activation in HUVECs by competing with activated Notch1 to bind to RBP-J. The analyses of biological functions showed that TAT-ANK promoted angiogenesis in vitro and in vivo by inhibiting Dll4-Notch1 signalling. CONCLUSIONS AND IMPLICATIONS: We synthesized and investigated the biological actions of TAT-ANK peptide, a new inhibitor of Notch signalling. This peptide will be of significant interest to research on Dll4-Notch1 signalling and to clinicians carrying out clinical trials using Notch signalling inhibitors. Furthermore, our findings will have important conceptual and therapeutic implications for angiogenesis-related diseases.


Subject(s)
Adaptor Proteins, Signal Transducing , Calcium-Binding Proteins , Neovascularization, Physiologic , Peptides , Receptor, Notch1 , Adaptor Proteins, Signal Transducing/metabolism , Animals , Calcium-Binding Proteins/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Neovascularization, Pathologic/drug therapy , Peptides/pharmacology , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction
6.
Mediators Inflamm ; 2021: 1791222, 2021.
Article in English | MEDLINE | ID: mdl-34646085

ABSTRACT

AIMS: Notch1 signaling regulates microglia activation, which promotes neuroinflammation. Neuroinflammation plays an essential role in various kinds of pain sensation, including bladder-related pain in bladder pain syndrome/interstitial cystitis (BPS/IC). However, the impact of Notch1 signaling on mechanical allodynia in cyclophosphamide- (CYP-) induced cystitis is unclear. This study is aimed at determining whether and how Notch1 signaling modulates mechanical allodynia of CYP-induced cystitis. METHODS: CYP was peritoneally injected to establish a bladder pain syndrome/interstitial cystitis (BPS/IC) rat model. A γ-secretase inhibitor, DAPT, was intrathecally injected to modulate Notch1 signaling indirectly. Mechanical withdrawal threshold in the lower abdomen was measured with von Frey filaments using the up-down method. The expression of Notch1 signaling, Iba-1, OX-42, TNF-α, and IL-1ß in the L6-S1 spinal dorsal horn (SDH) was measured with Western blotting analysis and immunofluorescence staining. RESULTS: Notch1 and Notch intracellular domain (NICD) were both upregulated in the SDH of the cystitis group. Moreover, the expression of Notch1 and NICD was negatively correlated with the mechanical withdrawal threshold of the cystitis rats. Furthermore, treatment with DAPT attenuated mechanical allodynia in CYP-induced cystitis and inhibited microglia activation, leading to decreased production of TNF-α and IL-1ß. CONCLUSION: Notch1 signaling contributes to mechanical allodynia associated with CYP-induced cystitis by promoting microglia activation and neuroinflammation. Our study showed that inhibition of Notch1 signaling might have therapeutic value for treating pain symptoms in BPS/IC.


Subject(s)
Cyclophosphamide/toxicity , Cystitis/physiopathology , Hyperalgesia/etiology , Microglia/physiology , Neuroinflammatory Diseases/etiology , Receptor, Notch1/physiology , Animals , Cystitis/chemically induced , Diamines/pharmacology , Female , Interleukin-1beta/biosynthesis , Rats , Rats, Sprague-Dawley , Receptor, Notch1/antagonists & inhibitors , Signal Transduction/physiology , Thiazoles/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis
7.
Cancer Med ; 10(11): 3689-3699, 2021 06.
Article in English | MEDLINE | ID: mdl-33960694

ABSTRACT

The marked overexpression of cyclin-dependent kinase 5 (CDK5) or Notch1 receptor, which plays critical roles in pancreatic ductal adenocarcinoma (PDAC) development, has been detected in numerous PDAC cell lines and tissues. Although, a previous study has demonstrated that CDK5 inhibition disrupts Notch1 functions in human umbilical vein endothelial cells, the mechanism underlying Notch1 activation regulated by CDK5 remains unclear. Herein, we identified a physical interaction between CDK5 and Notch1 in PDAC cells, with the Notch1 peptide phosphorylated by CDK5/p25 kinase. CDK5 blockade resulted in the profound inhibition of Notch signaling. Accordingly, CDK5 inhibition sensitized PDAC cell proliferation and migration following Notch inhibition. In conclusion, CDK5 positively regulates Notch1 function via phosphorylation, which in turn promotes cell proliferation and migration. The combinational inhibition of CDK5 and Notch signaling may be an effective strategy in the treatment of PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Cyclin-Dependent Kinase 5/metabolism , Pancreatic Neoplasms/metabolism , Receptor, Notch1/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Cyclin-Dependent Kinase 5/genetics , Dipeptides/pharmacology , Gene Silencing , Humans , Immunoprecipitation , Pancreatic Neoplasms/pathology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/metabolism , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Roscovitine/pharmacology , Signal Transduction
8.
Nat Commun ; 12(1): 2507, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33947863

ABSTRACT

Notch1 is a crucial oncogenic driver in T-cell acute lymphoblastic leukemia (T-ALL), making it an attractive therapeutic target. However, the success of targeted therapy using γ-secretase inhibitors (GSIs), small molecules blocking Notch cleavage and subsequent activation, has been limited due to development of resistance, thus restricting its clinical efficacy. Here, we systematically compare GSI resistant and sensitive cell states by quantitative mass spectrometry-based phosphoproteomics, using complementary models of resistance, including T-ALL patient-derived xenografts (PDX) models. Our datasets reveal common mechanisms of GSI resistance, including a distinct kinase signature that involves protein kinase C delta. We demonstrate that the PKC inhibitor sotrastaurin enhances the anti-leukemic activity of GSI in PDX models and completely abrogates the development of acquired GSI resistance in vitro. Overall, we highlight the potential of proteomics to dissect alterations in cellular signaling and identify druggable pathways in cancer.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Oligopeptides/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protein Kinase C/metabolism , Receptor, Notch1/antagonists & inhibitors , Acetophenones/pharmacology , Amyloid Precursor Protein Secretases/metabolism , Animals , Antineoplastic Agents/therapeutic use , Benzopyrans/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Chromatin Immunoprecipitation , Chromatography, High Pressure Liquid , Drug Resistance, Neoplasm/genetics , Gene Ontology , Humans , Inhibitory Concentration 50 , Mice , Mice, Inbred NOD , Phosphorylation , Protein Array Analysis , Protein Biosynthesis/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinases/metabolism , Proteomics , Receptor, Notch1/metabolism , Signal Transduction/drug effects , Tandem Mass Spectrometry , Xenograft Model Antitumor Assays
9.
Br J Haematol ; 194(1): 28-43, 2021 07.
Article in English | MEDLINE | ID: mdl-33942287

ABSTRACT

T-cell ALL (T-ALL) is an aggressive malignancy of T-cell progenitors. Although survival outcomes in T-ALL have greatly improved over the past 50 years, relapsed and refractory cases remain extremely challenging to treat and those who cannot tolerate intensive treatment continue to have poor outcomes. Furthermore, T-ALL has proven a more challenging immunotherapeutic target than B-ALL. In this review we explore our expanding knowledge of the basic biology of T-ALL and how this is paving the way for repurposing established treatments and the development of novel therapeutic approaches.


Subject(s)
Antineoplastic Agents/therapeutic use , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Antineoplastic Agents/administration & dosage , Antineoplastic Agents, Immunological/therapeutic use , Apoptosis/drug effects , Arabinonucleosides/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Cyclin-Dependent Kinases/antagonists & inhibitors , Genetic Heterogeneity , Humans , Immunotherapy , Immunotherapy, Adoptive , Janus Kinase Inhibitors/therapeutic use , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor, Notch1/antagonists & inhibitors , Receptors, Interleukin-7/antagonists & inhibitors , Salvage Therapy/methods , Signal Transduction/drug effects , Sulfonamides/therapeutic use , Therapies, Investigational/methods , Therapies, Investigational/trends , Treatment Outcome
10.
Cancer Res ; 81(12): 3347-3357, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33820800

ABSTRACT

In many human cancers, deregulation of the Notch pathway has been shown to play a role in the initiation and maintenance of the neoplastic phenotype. Aberrant Notch activity also plays a central role in the maintenance and survival of cancer stem cells (CSC), which underlie metastasis and resistance to therapy. For these reasons, inhibition of Notch signaling has become an exceedingly attractive target for cancer therapeutic development. However, attempts to develop Notch pathway-specific drugs have largely failed in the clinic, in part due to intestinal toxicity. Here, we report the discovery of NADI-351, the first specific small-molecule inhibitor of Notch1 transcriptional complexes. NADI-351 selectively disrupted Notch1 transcription complexes and reduced Notch1 recruitment to target genes. NADI-351 demonstrated robust antitumor activity without inducing intestinal toxicity in mouse models, and CSCs were ablated by NADI-351 treatment. Our study demonstrates that NADI-351 is an orally available and potent inhibitor of Notch1-mediated transcription that inhibits tumor growth with low toxicity, providing a potential therapeutic approach for improved cancer treatment. SIGNIFICANCE: This study showcases the first Notch1-selective inhibitor that suppresses tumor growth with limited toxicity by selectively ablating cancer stem cells.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , Esophageal Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Neoplastic Stem Cells/drug effects , Receptor, Notch1/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Apoptosis , Cell Proliferation , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Female , Humans , Mice , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Biochem Biophys Res Commun ; 554: 206-213, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33813076

ABSTRACT

Osteosarcoma is the most common primary bone tumor in children, teenagers and adolescents. Cancer stem cells (CSCs) have the function to self-renew and keep the phenotype of tumor, causing clinical treatment failure. Therefore, developing effective therapies to inhibit osteosarcoma progression is urgently necessary. Glycogen synthase kinase 3ß (GSK-3ß)is highly expressed in osteosarcoma. In the present study, we made an exploration on the anti-tumor effect of tideglusib (TID), a small-molecule inhibitor of GSK-3ß, and revealed the underlying mechanisms. Here, we found that TID markedly reduced the cell viability of different osteosarcoma cell lines. Cell cycle arrest distributed in G2/M was markedly up-regulated in TID-incubated osteosarcoma cells through enhancing p21 expression levels. Apoptosis was evidently induced in osteosarcoma cells via blocking Caspase-3 activation. Consistently, tumor growth was effectively suppressed in an established murine xenograft model with few toxicity and side effects in vivo. Furthermore, TID markedly repressed stem-cell-like activity in osteosarcoma cells through down-regulating NOTCH1 expression. Notably, rescuing NOTCH1 significantly abolished the role of TID in reducing cell proliferation and sarcosphere-formation. Mechanistically, we found that TID-inhibited NOTCH1 expression was associated with the blockage of AKT/GSK-3ß signaling pathway. In summary, we for the first time provided evidence that TID could effectively inhibit osteosarcoma progression through repressing cell proliferation, inducing apoptosis, suppressing stem-cell-like properties via down-regulating AKT/GSK-3ß/NOTCH1 signaling pathway. Thus, TID may be a promising therapeutic strategy for osteosarcoma treatment without side effects.


Subject(s)
Bone Neoplasms/drug therapy , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Osteosarcoma/drug therapy , Receptor, Notch1/antagonists & inhibitors , Stem Cells/drug effects , Thiadiazoles/pharmacology , Animals , Apoptosis/drug effects , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Humans , Mice , Mice, Inbred BALB C , Osteosarcoma/metabolism , Osteosarcoma/pathology , Stem Cells/pathology , Xenograft Model Antitumor Assays
12.
Cancer Sci ; 112(5): 1987-1996, 2021 May.
Article in English | MEDLINE | ID: mdl-33686706

ABSTRACT

Signaling pathways that drive bladder cancer (BC) progression may be promising and specific targets for systemic therapy. Here, we investigated the clinical significance and targetability of NOTCH and mitogen-activated protein kinase (MAPK) signaling for this aggressive malignancy. We assessed NOTCH1 and MAPK activity in 222 stage III and IV BC specimens of patients that had undergone radical cystectomy, and tested for clinical associations including cancer-specific and overall survival. We examined therapeutic effects of NOTCH and MAPK repression in a murine xenograft model of human bladder cancer cells and evaluated tumor growth and tumor cell plasticity. In BC, NOTCH1 and MAPK signaling marked two distinct tumor cell subpopulations. The combination of high NOTCH1 and high MAPK activity indicated poor cancer-specific and overall survival in univariate and multivariate analyses. Inhibition of NOTCH and MAPK in BC xenografts in vivo depleted targeted tumor cell subpopulations and revealed strong plasticity in signaling pathway activity. Combinatorial inhibition of NOTCH and MAPK signaling most strongly suppressed tumor growth. Our findings indicate that tumor cell subpopulations with high NOTCH and MAPK activity both contribute to tumor progression. Furthermore, we propose a new concept for BC therapy, which advocates specific and simultaneous targeting of these different tumor cell subpopulations through combined NOTCH and MAPK inhibition.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Receptor, Notch1/metabolism , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/metabolism , Aged , Analysis of Variance , Animals , Benzimidazoles/therapeutic use , Cell Line, Tumor , Dibenzazepines/therapeutic use , Disease Progression , Enzyme Inhibitors/therapeutic use , Female , Humans , Kaplan-Meier Estimate , Male , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Prognosis , Receptor, Notch1/antagonists & inhibitors , Regression Analysis , Signal Transduction , Tissue Array Analysis/methods , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
13.
Eur J Med Chem ; 216: 113247, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33652355

ABSTRACT

As a newly emerged technology, PROTAC (proteolysis targeting chimera) is a promising therapeutic strategy for varieties of diseases. Unlike small molecule inhibitors, PROTACs catalytically induce target proteins degradation, including currently "undruggable" target proteins. In addition, PROTACs can be a potentially successful strategy to overcome drug resistance. IAPs can inhibit apoptosis by inhibiting caspase, and also exhibits the activity of E3 ubiquitin ligase. Specific and nongenetic IAP-based protein erasers (SNIPERs) are hybrid molecules that designed based on IAPs, and used to degrade the target proteins closely associated with diseases. Their structures consist of three parts, including target protein ligand, E3 ligase ligand and the linker between them. SNIPERs (PROTACs) degrade diseases-associated proteins through human inherent ubiquitin-proteasome system. So far, many SNIPERs have been developed to treat diseases that difficult to handle by traditional methods, such as radiotherapy, chemotherapy and small molecule inhibitors, and showed promising prospects in application. In this paper, the recent advances of SNIPERs were summarized, and the chances and challenges associated with this area were also highlighted.


Subject(s)
Inhibitor of Apoptosis Proteins/metabolism , Ligands , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Fetal Proteins/antagonists & inhibitors , Fetal Proteins/metabolism , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/metabolism , Humans , Huntingtin Protein/antagonists & inhibitors , Huntingtin Protein/metabolism , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/metabolism , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/metabolism , Receptors, Androgen/chemistry , Receptors, Androgen/metabolism , Receptors, Estrogen/metabolism
14.
Mol Med Rep ; 23(5)2021 05.
Article in English | MEDLINE | ID: mdl-33649841

ABSTRACT

Recent studies have reported that gene amplified in squamous cell carcinoma 1 (GASC1) is involved in the progression of several types of cancer. However, whether GASC1 promotes glioma progression remains unknown. Therefore, the present study aimed to investigate the effect of GASC1 exposure on glioma tumorigenesis. The western blot demonstrated that grade III and IV glioma tissues exhibited a higher mRNA and protein expression of GASC1. Moreover, CD133+ U87 or U251 cells from magnetic cell separation exhibited a higher GASC1 expression. Invasion Transwell assay, clonogenic assay and wound healing assay have shown that GASC1 inhibition using a pharmacological inhibitor and specific short hairpin (sh)RNA suppressed the invasive, migratory and tumorsphere forming abilities of primary culture human glioma cells. Furthermore, GASC1­knockdown decreased notch receptor (Notch) responsive protein hes family bHLH transcription factor 1 (Hes1) signaling. GASC1 inhibition reduced notch receptor 1 (NOTCH1) expression, and a NOTCH1 inhibitor enhanced the effects of GASC1 inhibition on the CD133+ U87 or U251 cell tumorsphere forming ability, while NOTCH1 overexpression abrogated these effects. In addition, the GASC1 inhibitor caffeic acid and/or the NOTCH1 inhibitor DAPT (a γ­Secretase Inhibitor), efficiently suppressed the human glioma xenograft tumors. Thus, the present results demonstrated the importance of GASC1 in the progression of glioma and identified that GASC1 promotes glioma progression, at least in part, by enhancing NOTCH signaling, suggesting that GASC1/NOTCH1 signaling may be a potential therapeutic target for glioma treatment.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Receptor, Notch1/metabolism , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Caffeic Acids/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Survival/drug effects , Cell Survival/genetics , Diamines/pharmacology , Female , Gene Expression Regulation, Neoplastic , Glioma/drug therapy , Glioma/genetics , Humans , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Jumonji Domain-Containing Histone Demethylases/genetics , Male , Mice, Nude , RNA Interference , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Signal Transduction/genetics , Thiazoles/pharmacology , Xenograft Model Antitumor Assays/methods
15.
Cell Death Dis ; 12(4): 301, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33741899

ABSTRACT

The period circadian regulator 3 (PER3) has been reported to play a negative role in human immortalized bone marrow-derived Scp-1 cells (iBMSCs) and patient adipose-derived stromal cells (PASCs) or a negative/positive role in mice adipogenesis. However, human PER3 (hPER3) was identified as a positive regulator of human adipose tissue-derived stromal cells (hADSCs) adipogenesis in this study. Silencing or overexpression of hPER3 in hADSCs inhibited and promoted adipogenesis in vitro. In vivo, the overexpression of hPER3 increased high-fat diet-induced inguinal white adipose tissue (iWAT) and epididymal white adipose tissue (eWAT) forms, increasing systemic glucose intolerance and insulin resistance. Molecularly, hPER3 does not interact with hPPARγ, but represses Notch1 signaling pathway to enhance adipogenesis by interacting with hHSP90AA1, which is able to combine with the promoter of hNotch1 and inactivate its expression. Thus, our study revealed hPER3 as a critical positive regulator of hADSCs adipogenesis, which was different from the other types of cells, providing a critical role of it in treating obesity.


Subject(s)
Adipogenesis/physiology , Period Circadian Proteins/metabolism , Receptor, Notch1/antagonists & inhibitors , Animals , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Period Circadian Proteins/genetics , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Signal Transduction , Transfection
16.
Biochem Biophys Res Commun ; 547: 131-138, 2021 04 02.
Article in English | MEDLINE | ID: mdl-33610041

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) has become the most common cause of chronic liver disease worldwide and an urgent target for clinical intervention. Notch1 signaling pathway activity was found to be related to the severity of NAFLD, but the specific mechanism is not precise. Here, we investigated the potential mechanisms of Notch1 signaling in the development of NAFLD. Firstly, we found that Notch1 signaling is activated in free fatty acids-treated HepG2 cells accompanied by lipid accumulation, apoptosis, oxidative stress, and mitochondrial damage, which could be alleviated by Notch1 inhibitor N-[N-(3,5-Difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT). In the meantime, we found that administration of DAPT activated the autophagy pathway in NAFLD. Furthermore, the use of autophagy inhibitor chloroquine reversed the DAPT-mediated protective effect in NAFLD. All our results uncover a vital role of Notch1 in hepatocyte injury and metabolism of NAFLD, giving rise to a new sight for NAFLD treatment by regulation of Notch signaling and autophagy pathway.


Subject(s)
Dipeptides/pharmacology , Hepatocytes/pathology , Non-alcoholic Fatty Liver Disease/drug therapy , Receptor, Notch1/antagonists & inhibitors , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cells, Cultured , Disease Models, Animal , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Oxidative Stress/drug effects , Receptor, Notch1/metabolism , Signal Transduction
17.
Exp Neurol ; 339: 113645, 2021 05.
Article in English | MEDLINE | ID: mdl-33600815

ABSTRACT

Microglia are rapidly activated after acute ischemic stroke, and the polarization of microglial is associated with the prognosis of acute ischemic stroke. Lipoxin A4 (LXA4), an anti-inflammatory agent, has a protective effect against ischemic stroke. However, the role of LXA4 on the polarization of microglial after acute ischemic stroke remains undetermined. We hypothesized that LXA4 may exert the neuroprotective effect though regulating the polarization of microglial. In this study, clinical features of acute ischemic stroke were simulated using a rat model of model of middle cerebral artery occlusion (MCAO) in vivo and the BV2 microglia oxygen-glucose deprivation/reoxygenation model (OGD/R) in vitro. The protective effects of LXA4 on cerebral ischemia-reperfusion injury were determined using TTC staining, HE staining, and TUNEL staining. The expression of targeted genes was assayed using quantitative real-time PCR (qRT-PCR), immunofluorescence, and western blot to investigated the regulation of LXA4 on microglia polarization after acute ischemic stroke. We found that LXA4 exerted protective effects on focal cerebral ischemia-reperfusion injury and reduced the expression of the pro-inflammatory cytokines IL-1ß and TNF-α. Furthermore, LXA4 inhibited the expression of Notch-1, Hes1, iNOS and CD32 all of which are associated with the differentiation into M1 microglia. By contrast, LXA4 upregulated the expression of Hes5, Arg-1 and CD206 all of which are associated with M2 phenotype in microglia. In addition, blocking the Notch signaling pathway with the inhibitor DAPT significantly mitigated the effect of LXA4 on microglia differentiation. These data suggest that LXA4 may regulate the polarization of microglia after cerebral ischemia-reperfusion injury through the Notch signaling pathway.


Subject(s)
Brain Ischemia/drug therapy , Cell Polarity/drug effects , Microglia/drug effects , Receptor, Notch1/antagonists & inhibitors , Receptors, Lipoxin/administration & dosage , Reperfusion Injury/drug therapy , Animals , Brain Ischemia/metabolism , Cell Line , Cell Polarity/physiology , Male , Microglia/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Notch1/biosynthesis , Reperfusion Injury/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
18.
Life Sci ; 271: 119156, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33548289

ABSTRACT

Arecoline N-oxide (ANO), an oxidative metabolite of the areca nut, is a predictable initiator in carcinogenesis. The mechanisms of arecoline metabolites in human cancer specimens is still limited. This present study aims to estimate the oral squamous cell carcinoma (OSCC) inductive activity between arecoline metabolites in human cancer specimens/OSCC cells. We have collected 22 pairs (tumor and non-tumor part) of patient's specimens and checked for clinical characteristics. The identification of arecoline and its metabolites levels by using LC-MS/MS. The NOD/SCID mice model was used to check the OSCC inductive activity. The tumor part of OSCC samples exhibited higher levels of arecoline and ANO. Besides, ANO treated mice accelerates the NOTCH1, IL-17a and IL-1ß expressions compared to the control mice. ANO exhibited higher cytotoxicity, intracellular ROS levels and decline in antioxidant enzyme levels in OC-3 cells. The protein expression of NOTCH1 and proliferation marker levels are significantly lower in NOM treated cells. Overall, ANO induced initial stage carcinogenesis in the oral cavity via inflammation, ROS and depletion of antioxidant enzymes. Arecoline N-oxide mercapturic acid (NOM) attenuates the initiation of oral carcinogenesis.


Subject(s)
Acetylcysteine/therapeutic use , Arecoline/analogs & derivatives , Cyclic N-Oxides/toxicity , Free Radical Scavengers/therapeutic use , Mouth Neoplasms/chemically induced , Mouth Neoplasms/prevention & control , Adult , Animals , Arecoline/toxicity , Cells, Cultured , Female , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Mouth Neoplasms/metabolism , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/biosynthesis , Tumor Cells, Cultured
19.
Pharm Biol ; 59(1): 21-30, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33417512

ABSTRACT

CONTEXT: Berberine (BBR) is used to treat diarrhoea and gastroenteritis in the clinic. It was found to have anticolon cancer effects. OBJECTIVE: To study the anticolon cancer mechanism of BBR by connectivity map (CMAP) analysis. MATERIALS AND METHODS: CMAP based mechanistic prediction was conducted by comparing gene expression profiles of 10 µM BBR treated MCF-7 cells with that of clinical drugs such as helveticoside, ianatoside C, pyrvinium, gossypol and trifluoperazine. The treatment time was 12 h and two biological replications were performed. The DMSO-treated cells were selected as a control. The interaction between 100 µM BBR and target protein was measured by cellular thermal shift assay. The protein expression of 1-9 µM BBR treated SW480 cells were measured by WB assay. Apoptosis, cell cycle arrest, mitochondrial membrane potential (MMP) of 1-9 µM BBR treated SW480 cells were measured by flow cytometry and Hoechst 33342 staining methods. RESULTS: CMAP analysis found 14 Hsp90, HDAC, PI3K or mTOR protein inhibitors have similar functions with BBR. The experiments showed that BBR inhibited SW480 cells proliferation with IC50 of 3.436 µM, induced apoptosis, autophage, MMP depolarization and arrested G1 phase of cell cycle at 1.0 µM. BBR dose-dependently up-regulated PTEN, while inhibited Notch1, PI3K, Akt and mTOR proteins at 1.0-9.0 µM (p < 0.05). BBR also acted synergistically with Hsp90 and HDAC inhibitor (0.01 µM) in SW480 cells at 0.5 and 1.0 µM. DISCUSSION AND CONCLUSIONS: The integrative gene expression-based chemical genomic method using CMAP analysis may be applicable for mechanistic studies of other multi-targets drugs.


Subject(s)
Berberine/administration & dosage , Colonic Neoplasms/metabolism , PTEN Phosphohydrolase/biosynthesis , Phosphatidylinositol 3-Kinases/biosynthesis , Proto-Oncogene Proteins c-akt/biosynthesis , Receptor, Notch1/biosynthesis , TOR Serine-Threonine Kinases/biosynthesis , A549 Cells , Antineoplastic Agents/administration & dosage , Benzoquinones/administration & dosage , Colonic Neoplasms/drug therapy , Dose-Response Relationship, Drug , Drug Synergism , HCT116 Cells , Humans , Lactams, Macrocyclic/administration & dosage , MCF-7 Cells , Nylons , Phosphoinositide-3 Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrroles/administration & dosage , Receptor, Notch1/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , THP-1 Cells , TOR Serine-Threonine Kinases/antagonists & inhibitors
20.
Bosn J Basic Med Sci ; 21(2): 136-144, 2021 Apr 01.
Article in English | MEDLINE | ID: mdl-32415821

ABSTRACT

Childhood leukemia is cancer that seriously threatens the life of children in China. Poor sensitivity to chemotherapy and susceptibility to drug resistance are the reasons for the treatment of T-cell acute lymphocytic leukemia (T-ALL) being extremely difficult. Moreover, traditional intensive chemotherapy regimens cause great damage to children. Therefore, it is highly important to search for targeted drugs and develop a precise individualized treatment for child patients. There are activating mutations in the NOTCH1 gene in more than 50% of human T-ALLs and the Notch signaling pathway is involved in the pathogenesis of T-ALL. In this review, we summarize the progress in research on T-ALL and Notch1 signaling pathway inhibitors to provide a theoretical basis for the clinical treatment of T-ALL.


Subject(s)
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/physiology , Signal Transduction/physiology , Child , Humans , Receptor, Notch1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...