Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Dev Comp Immunol ; 125: 104147, 2021 12.
Article in English | MEDLINE | ID: mdl-34111502

ABSTRACT

In the Notch signaling pathway in vertebrates and invertebrates, the ligand Delta plays crucial roles in cell proliferation, differentiation, and immunity. Although the Notch signaling pathway has recently been implicated in the immune defense of Penaeus vannamei, the association of Delta with this immune response remains unclear. Here, we cloned and characterized the Delta homolog in P. vannamei (designated as PvDelta). PvDelta has a 2493 bp open reading frame (ORF) encoding a putative protein of 830 amino acids. Bioinformatics analysis revealed that PvDelta contains an N-terminal signal peptide, a conserved Notch ligand (MNNL) domain, a Delta/Serrate/Lag-2 segment, 9 epidermal growth factors segments, a transmembrane domain, and shares high homology with other Delta family members. Transcripts of PvDelta were detected in all shrimp tissues tested and were induced by Vibrio parahaemolyticus, white spot syndrome virus (WSSV), and lipopolysaccharide (LPS), indicating its involvement in shrimp immune response. Moreover, after PvDelta knockdown followed by LPS stimulation, the expression of Notch signaling pathway genes (i.e., PvNotch, PvCSL, and PvHey) was downregulated. Finally, shrimp depleted of PvDelta showed a lower survival rate in response to V. parahaemolyticus challenge. In sum, our data reveal that PvDelta is involved in the innate immunity of shrimp by positively modulating the Notch signaling pathway.


Subject(s)
Arthropod Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Penaeidae/immunology , Receptors, Notch/agonists , Vibrio Infections/immunology , Vibrio parahaemolyticus/physiology , Amino Acid Sequence , Animals , Arthropod Proteins/genetics , Cells, Cultured , Cloning, Molecular , Gene Knockdown Techniques , Immunity, Innate , Intracellular Signaling Peptides and Proteins/genetics , Ligands , Membrane Proteins/genetics , Sequence Alignment , Signal Transduction
2.
J Biol Chem ; 292(39): 16351-16359, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28842505

ABSTRACT

Glycosylation changes associated with cellular transformation can facilitate the growth and progression of tumors. Previously we discovered that the gene Mgat3 encoding the glycosyltransferase GnT-III is elevated in epithelial ovarian carcinomas (EOCs) and leads to the production of abnormal truncated N-linked glycan structures instead of the typical bisected forms. In this study, we are interested in discovering how these abnormal glycans impact the growth and progression of ovarian cancer. We have discovered using stable shRNA gene suppression that GnT-III expression controls the expansion of side-population cells, also known as cancer stem cells. More specifically, we found that GnT-III expression regulates the levels and activation of the heavily glycosylated Notch receptor involved in normal and malignant development. Suppression of GnT-III in EOC cell lines and primary tumor-derived cells resulted in an inhibition of Notch signaling that was more potent than pharmacologic blockage of Notch activation via γ-secretase inhibition. The inhibition resulted from the redirection of the Notch receptor to the lysosome, a novel mechanism. These findings demonstrate a new role for bisecting glycosylation in the control of Notch transport and demonstrate the therapeutic potential of inhibiting GnT-III as a treatment for controlling EOC growth and recurrence.


Subject(s)
Carcinoma/metabolism , N-Acetylglucosaminyltransferases/metabolism , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Ovarian Neoplasms/metabolism , Receptors, Notch/agonists , Signal Transduction , Animals , Carcinoma/pathology , Carcinoma/therapy , Cell Line, Tumor , Female , Glycosylation , Humans , Kaplan-Meier Estimate , Mice, Inbred NOD , N-Acetylglucosaminyltransferases/antagonists & inhibitors , N-Acetylglucosaminyltransferases/genetics , Neoplasm Invasiveness , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Ovary/metabolism , Ovary/pathology , Protein Processing, Post-Translational , RNA Interference , RNAi Therapeutics , Receptors, Notch/metabolism , Tissue Banks , Tumor Burden , Xenograft Model Antitumor Assays
3.
Nature ; 545(7654): 360-364, 2017 05 18.
Article in English | MEDLINE | ID: mdl-28489825

ABSTRACT

The Notch signalling pathway mediates cell fate decisions and is tumour suppressive or oncogenic depending on the context. During lung development, Notch pathway activation inhibits the differentiation of precursor cells to a neuroendocrine fate. In small-cell lung cancer, an aggressive neuroendocrine lung cancer, loss-of-function mutations in NOTCH genes and the inhibitory effects of ectopic Notch activation indicate that Notch signalling is tumour suppressive. Here we show that Notch signalling can be both tumour suppressive and pro-tumorigenic in small-cell lung cancer. Endogenous activation of the Notch pathway results in a neuroendocrine to non-neuroendocrine fate switch in 10-50% of tumour cells in a mouse model of small-cell lung cancer and in human tumours. This switch is mediated in part by Rest (also known as Nrsf), a transcriptional repressor that inhibits neuroendocrine gene expression. Non-neuroendocrine Notch-active small-cell lung cancer cells are slow growing, consistent with a tumour-suppressive role for Notch, but these cells are also relatively chemoresistant and provide trophic support to neuroendocrine tumour cells, consistent with a pro-tumorigenic role. Importantly, Notch blockade in combination with chemotherapy suppresses tumour growth and delays relapse in pre-clinical models. Thus, small-cell lung cancer tumours generate their own microenvironment via activation of Notch signalling in a subset of tumour cells, and the presence of these cells may serve as a biomarker for the use of Notch pathway inhibitors in combination with chemotherapy in select patients with small-cell lung cancer.


Subject(s)
Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Receptors, Notch/metabolism , Signal Transduction , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , Tumor Microenvironment , Animals , Cell Differentiation , Cell Proliferation/drug effects , Disease Models, Animal , Female , Humans , Lung Neoplasms/drug therapy , Male , Mice , Neoplasm Recurrence, Local/prevention & control , Receptors, Notch/agonists , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/deficiency , Repressor Proteins/metabolism , Small Cell Lung Carcinoma/drug therapy
4.
Biochem Pharmacol ; 118: 18-30, 2016 Oct 15.
Article in English | MEDLINE | ID: mdl-27520484

ABSTRACT

It is believed that tumor cells can give rise to endothelial cells and tumor endothelium has a neoplastic origin. Yet, the stimuli and underlying mechanism remain unclear. Here, we demonstrate that adriamycin or paclitaxel, first-line chemotherapy agent, induced breast cancer cells to generate morphological, phenotypical and functional features of endothelial cells in vitro. In xenografts models, challenges from adriamycin or paclitaxel induced cancer cells to generate the majority of microvessels. Importantly, in breast cancer specimens from patients with neoadjuvant anthracycline-based or taxane-based chemotherapy, tumor-derived endothelial microvessels, lined by EGFR-amplified or/and TP53+-CD31+ endothelial cells, was significantly higher in patients with progressive or stable disease (PD/SD) than in those with a partial or complete response (PR/CR). Further, exposure to the Notch signaling inhibitor and gene silencing studies showed that Notch signaling inhibition or silencing Nothc4/Dll3 decreased endothelial markers and function of tumor-derived endothelial cells under chemotherapy treatment, which may be through VEGFR3. Thus, our findings demonstrate that chemotherapy induces functional tumor-derived endothelial microvessels by mediating Notch signaling and VEGF signaling, and may provide new targets for anti-angiogenesis therapy in breast cancer.


Subject(s)
Antineoplastic Agents/adverse effects , Breast Neoplasms/drug therapy , Endothelium, Vascular/drug effects , Neoplasm Proteins/agonists , Neovascularization, Pathologic/chemically induced , Receptors, Notch/agonists , Signal Transduction/drug effects , Animals , Anthracyclines/adverse effects , Anthracyclines/therapeutic use , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/toxicity , Biomarkers/metabolism , Breast Neoplasms/blood supply , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Bridged-Ring Compounds/adverse effects , Bridged-Ring Compounds/therapeutic use , Doxorubicin/adverse effects , Doxorubicin/therapeutic use , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Humans , MCF-7 Cells , Mice, Nude , Microvessels/drug effects , Microvessels/metabolism , Microvessels/pathology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Paclitaxel/adverse effects , Paclitaxel/therapeutic use , RNA Interference , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/genetics , Receptors, Notch/metabolism , Specific Pathogen-Free Organisms , Taxoids/adverse effects , Taxoids/therapeutic use , Xenograft Model Antitumor Assays
5.
Cancer Res ; 75(22): 4728-41, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26404003

ABSTRACT

Activation of Notch signaling in hematopoietic cells by tumors contributes to immune escape. T-cell defects in tumors can be reversed by treating tumor-bearing mice with multivalent forms of the Notch receptor ligand DLL-1, but the immunologic correlates of this effect have not been elucidated. Here, we report mechanistic insights along with the efficacy of combinational treatments of multivalent DLL-1 with oncoprotein targeting drugs in preclinical mouse models of lung cancer. Systemic DLL-1 administration increased T-cell infiltration into tumors and elevated numbers of CD44(+)CD62L(+)CD8(+) memory T cells while decreasing the number of regulatory T cells and limiting tumor vascularization. This treatment was associated with upregulation of Notch and its ligands in tumor-infiltrating T cells enhanced expression of T-bet and phosphorylation of Stat1/2. Adoptive transfer of T cells from DLL1-treated tumor-bearing immunocompetent hosts into tumor-bearing SCID-NOD immunocompromised mice attenuated tumor growth and extended tumor-free survival in the recipients. When combined with the EGFR-targeted drug erlotinib, DLL-1 significantly improved progression-free survival by inducing robust tumor-specific T-cell immunity. In tissue culture, DLL1 induced proliferation of human peripheral T cells, but lacked proliferative or clonogenic effects on lung cancer cells. Our findings offer preclinical mechanistic support for the development of multivalent DLL1 to stimulate antitumor immunity.


Subject(s)
Intercellular Signaling Peptides and Proteins/pharmacology , Lung Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Antineoplastic Agents/pharmacology , Blotting, Western , Calcium-Binding Proteins , Cell Line, Tumor , Disease Models, Animal , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride/pharmacology , Female , Flow Cytometry , Humans , Immunohistochemistry , Immunotherapy/methods , Lung Neoplasms/drug therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Receptors, Notch/agonists , Recombinant Fusion Proteins/pharmacology
6.
Adv Exp Med Biol ; 804: 67-92, 2014.
Article in English | MEDLINE | ID: mdl-24924169

ABSTRACT

The Notch pathway has been described as an oncogene in osteosarcoma, but the myriad functions of all the members of this complex signaling pathway, both in malignant cells and nonmalignant components of tumors, make it more difficult to define Notch as simply an oncogene or a tumor suppressor. The cell-autonomous behaviors caused by Notch pathway manipulation may vary between cell lines but can include changes in proliferation, migration, invasiveness, oxidative stress resistance, and expression of markers associated with stemness or tumor-initiating cells. Beyond these roles, Notch signaling also plays a vital role in regulating tumor angiogenesis and vasculogenesis, which are vital aspects of osteosarcoma growth and behavior in vivo. Further, osteosarcoma cells themselves express relatively low levels of Notch ligand, making it likely that nonmalignant cells, especially endothelial cells and pericytes, are the major source of Notch activation in osteosarcoma tumors in vivo and in patients. As a result, Notch pathway expression is not expected to be uniform across a tumor but likely to be highest in those areas immediately adjacent to blood vessels. Therapeutic targeting of the Notch pathway is likewise expected to be complicated. Most pharmacologic approaches thus far have focused on inhibition of gamma secretase, a protease of the presenilin complex. This enzyme, however, has numerous other target proteins that would be expected to affect osteosarcoma behavior, including CD44, the WNT/ß-catenin pathway, and Her-4. In addition, Notch plays a vital role in tissue and organ homeostasis in numerous systems, and toxicities, especially GI intolerance, have limited the effectiveness of gamma secretase inhibitors. New approaches are in development, and the downstream targets of Notch pathway signaling also may turn out to be good targets for therapy. In summary, a full understanding of the complex functions of Notch in osteosarcoma is only now unfolding, and this deeper knowledge will help position the field to better utilize novel therapies as they are developed.


Subject(s)
Bone Neoplasms/blood supply , Gene Expression Regulation, Neoplastic , Lung Neoplasms/blood supply , Osteosarcoma/blood supply , Receptors, Notch/genetics , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Antineoplastic Agents/therapeutic use , Bone Neoplasms/drug therapy , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Enzyme Inhibitors/therapeutic use , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Neovascularization, Pathologic , Osteosarcoma/drug therapy , Osteosarcoma/genetics , Osteosarcoma/secondary , Receptors, Notch/agonists , Receptors, Notch/antagonists & inhibitors , Signal Transduction
7.
Vitam Horm ; 95: 391-405, 2014.
Article in English | MEDLINE | ID: mdl-24559926

ABSTRACT

Beta-cell replacement represents the optimal therapy for type 1 diabetes. Efforts to manipulate ß-cell proliferation and differentiation could be advanced by a better understanding of the normal pathways regulating ß-cell development and renewal. NOTCH signaling is a highly conserved pathway which plays a central role in pancreas development. Cell-lineage tracing has revealed the reactivation of the NOTCH pathway in adult human ß cells cultured under conditions which induce cell proliferation and dedifferentiation. Inhibition of NOTCH signaling in dedifferentiated cells following ex vivo expansion has been shown to promote restoration of the ß-cell phenotype. This approach may increase the availability of functional ß cells for transplantation.


Subject(s)
Insulin-Secreting Cells/cytology , Models, Biological , Receptors, Notch/metabolism , Signal Transduction , Animals , Cell Dedifferentiation , Cell Differentiation , Cells, Cultured , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Diabetes Mellitus/therapy , Humans , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/transplantation , Ligands , Protein Isoforms/agonists , Protein Isoforms/metabolism , Receptors, Notch/agonists
8.
Am J Physiol Cell Physiol ; 306(9): C871-8, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24573085

ABSTRACT

Notch signaling plays a critical role in controlling proliferation and differentiation of pulmonary arterial smooth muscle cells (PASMC). Upregulated Notch ligands and Notch3 receptors in PASMC have been reported to promote the development of pulmonary vascular remodeling in patients with pulmonary arterial hypertension (PAH) and in animals with experimental pulmonary hypertension. Activation of Notch receptors by their ligands leads to the cleavage of the Notch intracellular domain (NICD) to the cytosol by γ-secretase; NICD then translocates into the nucleus to regulate gene transcription. In this study, we examined whether short-term activation of Notch functionally regulates store-operated Ca(2+) entry (SOCE) in human PASMC. Treatment of PASMC with the active fragment of human Jagged-1 protein (Jag-1) for 15-60 min significantly increased the amplitude of SOCE induced by passive deletion of Ca(2+) from the intracellular stores, the sarcoplasmic reticulum (SR). The Jag-1-induced enhancement of SOCE was time dependent: the amplitude was maximized at 30 min of treatment with Jag-1, which was closely correlated with the time course of Jag-1-mediated increase in NICD protein level. The scrambled peptide of Jag-1 active fragment had no effect on SOCE. Inhibition of γ-secretase by N-[N-(3,5-difluorophenacetyl-L-alanyl)]-S-phenylglycine t-butyl ester (DAPT) significantly attenuated the Jag-1-induced augmentation of SOCE. In addition to the short-term effect, prolonged treatment of PASMC with Jag-1 for 48 h also markedly enhanced the amplitude of SOCE. These data demonstrate that short-term activation of Notch signaling enhances SOCE in PASMC; the NICD-mediated functional interaction with store-operated Ca(2+) channels (SOC) may be involved in the Jag-1-mediated enhancement of SOCE in human PASMC.


Subject(s)
Calcium Channel Agonists/pharmacology , Calcium Channels/drug effects , Calcium Signaling/drug effects , Calcium-Binding Proteins/pharmacology , Intercellular Signaling Peptides and Proteins/pharmacology , Membrane Proteins/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Peptide Fragments/pharmacology , Receptors, Notch/agonists , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Animals , Calcium Channels/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Humans , Jagged-1 Protein , Male , Mice , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Receptors, Notch/metabolism , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Serrate-Jagged Proteins , Time Factors
9.
ACS Appl Mater Interfaces ; 6(3): 1652-61, 2014 Feb 12.
Article in English | MEDLINE | ID: mdl-24405311

ABSTRACT

Engineered scaffold surface provides stem cells with vital cues that could determine the eventual fate of stem cells. In this work, biodegradable poly(L-lactide-co-ε-caprolactone) (PLCL) scaffold conjugated with Notch agonist-Jagged-1(JAG) peptide (2.1 kDa) was prepared to initiate myogenic differentiation of human mesenchymal stem cells (hMSCs). The scaffold surface was activated with oxygen plasma and acrylic acid was engrafted via UV polymerization to form a surface bearing carboxylic groups. JAG peptide was subsequently immobilized onto the carboxylated scaffold surface. Surface chemistry and topography were examined using attenuated total reflection Fourier transform infrared, X-ray photoelectron spectroscopy, and atomic force microscopy. Quantitative real time polymerase chain reaction analysis revealed activation of the Notch pathway; furthermore, several specific markers associated with myogenic but not osteogenic differentiation were shown to be up-regulated in hMSCs cultured on the engineered surface. The pro-myocardial effect of surface bound JAG peptide was further affirmed via immunodetection of the distinct myocardial marker, cardiac troponin T. Collectively, our results suggest that PLCL conjugated JAG peptide is a viable strategy to enhance the functional potential of scaffolds to be used as a bioengineered cardiac patch in myocardial infarction repair.


Subject(s)
Biocompatible Materials/pharmacology , Calcium-Binding Proteins/pharmacology , Cell Differentiation/drug effects , Intercellular Signaling Peptides and Proteins/pharmacology , Membrane Proteins/pharmacology , Mesenchymal Stem Cells/cytology , Muscle Development/drug effects , Receptors, Notch/agonists , Tissue Scaffolds/chemistry , Cell Differentiation/genetics , Cells, Cultured , Free Radicals/analysis , Gene Expression Regulation/drug effects , Humans , Jagged-1 Protein , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Microscopy, Atomic Force , Muscle Development/genetics , Peptides/pharmacology , Photoelectron Spectroscopy , Polyesters/pharmacology , Serrate-Jagged Proteins , Signal Transduction/drug effects , Spectroscopy, Fourier Transform Infrared , Surface Properties
10.
Nat Methods ; 10(8): 696-7, 2013 Aug.
Article in English | MEDLINE | ID: mdl-24058977
11.
Science ; 340(6135): 991-4, 2013 May 24.
Article in English | MEDLINE | ID: mdl-23704575

ABSTRACT

Cell-cell and cell-matrix mechanical interactions through membrane receptors direct a wide range of cellular functions and orchestrate the development of multicellular organisms. To define the single molecular forces required to activate signaling through a ligand-receptor bond, we developed the tension gauge tether (TGT) approach in which the ligand is immobilized to a surface through a rupturable tether before receptor engagement. TGT serves as an autonomous gauge to restrict the receptor-ligand tension. Using a range of tethers with tunable tension tolerances, we show that cells apply a universal peak tension of about 40 piconewtons (pN) to single integrin-ligand bonds during initial adhesion. We find that less than 12 pN is required to activate Notch receptors. TGT can also provide a defined molecular mechanical cue to regulate cellular functions.


Subject(s)
Cell Communication , Integrins/agonists , Mechanotransduction, Cellular , Receptors, Notch/agonists , Animals , CHO Cells , Cell Adhesion , Cricetinae , Cricetulus , DNA/chemistry , HEK293 Cells , Humans , Ligands , Mice , NIH 3T3 Cells , Shear Strength , Surface Properties
12.
J Exp Med ; 210(2): 321-37, 2013 Feb 11.
Article in English | MEDLINE | ID: mdl-23359069

ABSTRACT

Although aberrant Notch activation contributes to leukemogenesis in T cells, its role in acute myelogenous leukemia (AML) remains unclear. Here, we report that human AML samples have robust expression of Notch receptors; however, Notch receptor activation and expression of downstream Notch targets are remarkably low, suggesting that Notch is present but not constitutively activated in human AML. The functional role of these Notch receptors in AML is not known. Induced activation through any of the Notch receptors (Notch1-4), or through the Notch target Hairy/Enhancer of Split 1 (HES1), consistently leads to AML growth arrest and caspase-dependent apoptosis, which are associated with B cell lymphoma 2 (BCL2) loss and enhanced p53/p21 expression. These effects were dependent on the HES1 repressor domain and were rescued through reexpression of BCL2. Importantly, activated Notch1, Notch2, and HES1 all led to inhibited AML growth in vivo, and Notch inhibition via dnMAML enhanced proliferation in vivo, thus revealing the physiological inhibition of AML growth in vivo in response to Notch signaling. As a novel therapeutic approach, we used a Notch agonist peptide that led to significant apoptosis in AML patient samples. In conclusion, we report consistent Notch-mediated growth arrest and apoptosis in human AML, and propose the development of Notch agonists as a potential therapeutic approach in AML.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Receptors, Notch/metabolism , Adolescent , Animals , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Survival , Child , DNA-Binding Proteins/genetics , Gene Expression , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Infant , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mutation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Receptors, Notch/agonists , Receptors, Notch/genetics , Signal Transduction , Transcription Factor HES-1 , Transcription Factors/genetics , Tumor Suppressor Protein p53/metabolism
13.
J Exp Med ; 210(2): 301-19, 2013 Feb 11.
Article in English | MEDLINE | ID: mdl-23359070

ABSTRACT

Notch signaling pathway activation is known to contribute to the pathogenesis of a spectrum of human malignancies, including T cell leukemia. However, recent studies have implicated the Notch pathway as a tumor suppressor in myeloproliferative neoplasms and several solid tumors. Here we report a novel tumor suppressor role for Notch signaling in acute myeloid leukemia (AML) and demonstrate that Notch pathway activation could represent a therapeutic strategy in this disease. We show that Notch signaling is silenced in human AML samples, as well as in AML-initiating cells in an animal model of the disease. In vivo activation of Notch signaling using genetic Notch gain of function models or in vitro using synthetic Notch ligand induces rapid cell cycle arrest, differentiation, and apoptosis of AML-initiating cells. Moreover, we demonstrate that Notch inactivation cooperates in vivo with loss of the myeloid tumor suppressor Tet2 to induce AML-like disease. These data demonstrate a novel tumor suppressor role for Notch signaling in AML and elucidate the potential therapeutic use of Notch receptor agonists in the treatment of this devastating leukemia.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Receptors, Notch/metabolism , Animals , Cell Differentiation , Cell Survival , DNA-Binding Proteins/metabolism , Dioxygenases , Disease Models, Animal , Gene Silencing , Homeostasis , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Ligands , Mice , Mice, Transgenic , Mutation , Proto-Oncogene Proteins/metabolism , Receptors, Notch/agonists , Signal Transduction , Tumor Suppressor Proteins/metabolism
14.
Biochem Biophys Res Commun ; 428(4): 518-24, 2012 Nov 30.
Article in English | MEDLINE | ID: mdl-23111325

ABSTRACT

One of the greatest challenges in multiple myeloma (MM) treatment is to overcome drug resistance. Many pathways are involved including Notch signaling. Notch receptors are expressed by MM cells and Notch ligand Dll1 is present on bone marrow (BM) stromal cells. In this study, we demonstrate that Dll1 can activate Notch signaling mostly through Notch2 receptor and can contribute to drug resistance to bortezomib, both in murine and human MM cells. Blocking the Notch pathway by DAPT (gamma secretase inhibitor) could reverse this effect and increased sensitivity to bortezomib. We describe the upregulation of CYP1A1, a Cytochrome P450 enzyme involved in drug metabolism, as a possible mechanism of Dll1/Notch induced bortezomib resistance. This was confirmed by inhibition experiments using α-Naphthoflavone or CYP1A1-siRNA that resulted in an increased sensitivity to bortezomib. In addition, in vivo data showed that combination treatment of DAPT with bortezomib was able to increase bortezomib sensitivity and prolonged overall survival in the 5T33MM mouse model. Our data provide a potential strategy to overcome bortezomib resistance by Notch inhibition in MM therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Cytochrome P-450 CYP1A1/biosynthesis , Drug Resistance, Neoplasm , Intercellular Signaling Peptides and Proteins/agonists , Membrane Proteins/agonists , Multiple Myeloma/metabolism , Pyrazines/pharmacology , Receptors, Notch/agonists , Animals , Benzoflavones/pharmacology , Bortezomib , Calcium-Binding Proteins , Cell Line, Tumor , Cytochrome P-450 CYP1A1/antagonists & inhibitors , Cytochrome P-450 CYP1A1/genetics , Humans , Mice , Up-Regulation
15.
Circulation ; 124(11 Suppl): S197-203, 2011 Sep 13.
Article in English | MEDLINE | ID: mdl-21911813

ABSTRACT

BACKGROUND: We investigated the effect of epidermal growth factor-like domain 7 (Egfl7) on nuclear factor-κB activation, intercellular adhesion molecule-1 expression, and neutrophil adhesion to human coronary artery endothelial cells after calcineurin-inhibition-induced injury. METHODS AND RESULTS: Human coronary endothelial cells were incubated with cyclosporine (CyA) 10 µg/mL with or without Egfl7 (100 ng/mL) or the Notch receptor activator Jagged1 (200 ng/mL) for 6 to 48 hours. CyA upregulated nuclear factor-κB (p65) activity (128 ± 2% of control, P<0.001) in nuclear extracts, as determined with a DNA-binding activity ELISA. This activity was inhibited by Egfl7 (86 ± 3% of control; P<0.001 versus CyA alone). Jagged1 blocked Egfl7-induced nuclear factor-κB inhibition (105 ± 4% of control; P<0.05 versus CyA plus Egfl7). CyA upregulated cell-surface intercellular adhesion molecule-1 expression (215 ± 13% of control; P<0.001), as determined by flow cytometry. This expression was suppressed by Egfl7 (148 ± 5%; P<0.001 versus CyA alone). Jagged1 attenuated the intercellular adhesion molecule-1-suppressive effect of Egfl7 when administered with CyA (193 ± 3% versus 148 ± 5%; P<0.01). CyA increased neutrophil adhesion to human coronary endothelial cells (control 20 ± 5%, CyA 37 ± 3%; P<0.001 versus control) in a nonstatic neutrophil adhesion assay. This increase was attenuated by Egfl7 (22 ± 6%; P<0.001 versus CyA alone). Jagged 1 attenuated the effect of Egfl7 on neutrophil adhesion (31±3%; P<0.001 versus Egfl7 plus CyA). CONCLUSIONS: Our study reveals that Egfl7 is a potent inhibitor of neutrophil adhesion to human coronary endothelial cells subsequent to calcineurin-inhibition-induced injury. Mechanistically, Egfl7 blocked nuclear factor-κB pathway activation and intercellular adhesion molecule-1 expression, which suggests that it may have significant antiinflammatory properties. Because Jagged1 blocked the effect of Egfl7, Notch receptor antagonism may contribute to the mechanism of action of Egfl7.


Subject(s)
Calcineurin Inhibitors , Coronary Vessels/cytology , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Neutrophils/cytology , Neutrophils/drug effects , Calcineurin/drug effects , Calcium-Binding Proteins/pharmacology , Cell Adhesion/drug effects , Cells, Cultured , Cyclosporine/pharmacology , EGF Family of Proteins , Endothelium, Vascular/metabolism , Humans , Immunosuppressive Agents/pharmacology , Intercellular Adhesion Molecule-1/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Jagged-1 Protein , Membrane Proteins/pharmacology , NF-kappa B/metabolism , Nitric Oxide/metabolism , Receptors, Notch/agonists , Serrate-Jagged Proteins , Signal Transduction/drug effects , Signal Transduction/physiology
16.
Neuroscience ; 154(3): 946-52, 2008 Jun 26.
Article in English | MEDLINE | ID: mdl-18495362

ABSTRACT

The appropriate level of microtubule stability is fundamental in neurons to assure correct polarity, migration, vesicles transport and to prevent axonal degeneration. In the present study, we have identified Notch pathway as an endogenous microtubule stabilizer. Stimulation of Notch receptors by exposure of mouse cortical neurons to the Notch ligand Jagged1 resulted in increased microtubule stability, as measured by using antibodies against post-translationally modified alpha tubulin, and changes in axonal morphology and branching, with varicosity loss, thicker neurites and enlarged growth cones. Similar effects were found after exposure of the cells to different doses of Taxol. However, contrary to Taxol, Jagged1 induced downregulation of the microtubule severing protein Spastin. We suggest that a fine-tuned manipulation of Notch signaling may represent a novel approach to modulate neuronal cytoskeleton plasticity.


Subject(s)
Cerebral Cortex/physiology , Microtubules/drug effects , Neurons/drug effects , Receptors, Notch/agonists , Adenosine Triphosphatases/biosynthesis , Adenosine Triphosphatases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/physiology , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/physiology , Jagged-1 Protein , Ligands , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Microscopy, Confocal , Paclitaxel/pharmacology , Polyglutamic Acid/metabolism , RNA/biosynthesis , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Serrate-Jagged Proteins , Signal Transduction/drug effects , Spastin , Tubulin/metabolism , Tyrosine/metabolism
17.
J Immunol ; 180(3): 1598-608, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18209056

ABSTRACT

The mechanisms underlying the immunomodulatory functions of mesenchymal stem cells (MSC) on dendritic cells (DC) have been shown to involve soluble factors, such as IL-6 or TGF-beta, or cell-cell contact, or both depending on the report referenced. In this study, we intend to clarify these mechanisms by examining the immunosuppressive effect of human adult MSC on adult DC differentiated from CD34(+) hemopoietic progenitor cells (HPC). MSC have been shown to inhibit interstitial DC differentiation from monocytes and umbilical CD34(+) HPC. In this study, we confirm that MSC not only halt interstitial DC but also Langerhans cell differentiation from adult CD34(+) HPC, as assessed by the decreased expression of CD1a, CD14, CD86, CD80, and CD83 Ags on their cell surface. Accordingly, the functional capacity of CD34(+) HPC-derived DC (CD34-DC) to stimulate alloreactive T cells was impaired. Furthermore, we showed that 1) MSC inhibited commitment of CD34(+) HPC into immature DC, but not maturation of CD34-DC, 2) this inhibitory effect was reversible, and 3) DC generated in coculture with MSC (MSC-DC) induced the generation of alloantigen-specific regulatory T cells following secondary allostimulation. Conditioned medium from MSC cultures showed some inhibitory effect independent of IL-6, M-CSF, and TGF-beta. In comparison, direct coculture of MSC with CD34(+) HPC resulted in much stronger immunosuppressive effect and led to an activation of the Notch pathway as assessed by the overexpression of Hes1 in MSC-DC. Finally, DAPT, a gamma-secretase inhibitor that inhibits Notch signaling, was able to overcome MSC-DC defects. In conclusion, our data suggest that MSC license adult CD34(+) HPC to differentiate into regulatory DC through activation of the Notch pathway.


Subject(s)
Cell Differentiation , Dendritic Cells/immunology , Hematopoietic Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Receptors, Notch/agonists , Antibodies, Monoclonal/pharmacology , Antigens, CD34/analysis , CD28 Antigens/immunology , CD3 Complex/immunology , Cells, Cultured , Coculture Techniques , Cross-Priming , Dendritic Cells/cytology , Humans , T-Lymphocytes/drug effects
18.
Cell Tissue Res ; 332(1): 25-35, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18196275

ABSTRACT

A number of signaling molecules and transcription factors play important roles in the development of the autonomic nervous system. Here, we show that mouse trunk neural crest cells can differentiate into autonomic neurons expressing mammalian achaete-scute homolog 1 (mash1), Phox2b, tyrosine hydroxylase, and/or dopamine-beta-hydroxylase in the absence of bone morphogenetic protein (BMP)-4. The expression of mash1 and Phox2b is induced even in the presence of noggin or chordin, which are inhibitors of BMP signaling. Whereas these autonomic neurons do not express c-ret, the receptor for glial-cell-line-derived neurotrophic factor (GDNF), GDNF promotes the differentiation of c-ret-positive autonomic neurons in the presence of noggin. Autonomic neurogenesis is completely prevented by fibroblast growth factor (FGF)-2 treatment or by activation of Notch signaling. Furthermore, the suppression of Phox2b expression by FGF-2 can be recovered by treatment with Notch-1 small interfering RNA. Our data suggest that BMP-independent mechanisms promote the differentiation of autonomic neurons, and that FGF-2 suppresses autonomic neurogenesis by means of the activation of Notch signaling.


Subject(s)
Bone Morphogenetic Proteins/physiology , Cell Differentiation/physiology , Neurons/cytology , Signal Transduction/physiology , Animals , Autonomic Pathways/cytology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/pharmacology , Carrier Proteins/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chick Embryo , Dopamine beta-Hydroxylase/metabolism , Fibroblast Growth Factor 2/pharmacology , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Glycoproteins/pharmacology , Homeodomain Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , Neural Crest/cytology , Neurons/drug effects , Neurons/metabolism , Proto-Oncogene Proteins c-ret/metabolism , RNA, Small Interfering/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Receptors, Notch/agonists , Signal Transduction/drug effects , Transcription Factor HES-1 , Transcription Factors/metabolism , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/pharmacology , Tyrosine 3-Monooxygenase/metabolism
19.
Proc Natl Acad Sci U S A ; 104(49): 19327-32, 2007 Dec 04.
Article in English | MEDLINE | ID: mdl-18042722

ABSTRACT

Acinar-to-ductal metaplasia in the pancreas is associated with an increased risk for tumorigenesis. Molecular dissection of this process in vitro has shown that primary acinar cells, in response to EGF receptor ligands, can transdifferentiate into duct-like epithelia, passing through a nestin-positive intermediate, in a Notch pathway-dependent manner. Here, we show that in vitro acinar transdifferentiation depends on matrix metalloproteinase 7 (MMP-7), a proteinase expressed in most metaplastic epithelia in vivo. MMP-7 was found to be required for Notch activation, which leads to dedifferentiation of acinar cells to the nestin-positive transitional cell. Besides being necessary for acinar transdifferentiation, it was found that MMP-7 activity was sufficient to induce the process, indicating that molecular signals capable of initiating MMP-7 expression also have the potential to induce formation of metaplastic epithelia in the pancreas.


Subject(s)
Adenocarcinoma/pathology , Carcinoma, Acinar Cell/pathology , Carcinoma, Pancreatic Ductal/pathology , Cell Transdifferentiation , Matrix Metalloproteinase 7/metabolism , Pancreatic Neoplasms/pathology , Receptors, Notch/agonists , Adenocarcinoma/enzymology , Animals , Carcinoma, Acinar Cell/enzymology , Carcinoma, Pancreatic Ductal/enzymology , Cells, Cultured , Intermediate Filament Proteins/analysis , Matrix Metalloproteinase 7/genetics , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/analysis , Nestin , Pancreatic Neoplasms/enzymology
20.
Development ; 134(6): 1161-9, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17332535

ABSTRACT

The specification of polar, main-body and stalk follicle cells in the germarium of the Drosophila ovary plays a key role in the formation of the egg chamber and polarisation of its anterior-posterior axis. High levels of Notch pathway activation, resulting from a germline Delta ligand signal, induce polar cells. Here we show that low Notch activation levels, originating from Delta expressed in the polar follicle cells, are required for stalk formation. The metalloprotease Kuzbanian-like, which cleaves and inactivates Delta, reduces the level of Delta signaling between follicle cells, thereby limiting the size of the stalk. We find that Notch activation is required in a continuous fashion to maintain the polar and stalk cell fates. We further demonstrate that mutual antagonism between the Notch and JAK/STAT signaling pathways provides a crucial facet of follicle cell patterning. Notch signaling in polar and main-body follicle cells inhibits JAK/STAT signaling by preventing STAT nuclear translocation, thereby restricting the influence of this pathway to stalk cells. Conversely, signaling by JAK/STAT reduces Notch signaling in the stalk. Thus, variations in the levels of Notch pathway activation, coupled with a continuous balance between the Notch and JAK/STAT pathways, specify the identity of the different follicle cell types and help establish the polarity of the egg chamber.


Subject(s)
Drosophila/growth & development , Janus Kinases/metabolism , Oogenesis , Ovarian Follicle/growth & development , Receptors, Notch/agonists , STAT Transcription Factors/metabolism , Animals , Body Patterning , Cell Polarity , Drosophila/genetics , Drosophila/metabolism , Female , Janus Kinases/antagonists & inhibitors , Oogenesis/genetics , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , STAT Transcription Factors/antagonists & inhibitors , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...