Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cytokine ; 125: 154834, 2020 01.
Article in English | MEDLINE | ID: mdl-31491724

ABSTRACT

Sjögren's syndrome (SS) is a systemic autoimmune disease with severe dysfunction of glandular secretory function mediated by T and B lymphocyte infiltration into the exocrine glands, including the salivary and lacrimal glands. Follicular helper T (Tfh) cells exacerbate the disease by causing B cell hyperactivity. Inhibitor of DNA binding 3 (Id3) deficiency causes activation of Tfh cells and is known to be a clinical manifestation of human SS disease. In this study, we investigated the mechanism of action of Pax3, which is reduced in SS and can interact with Id3, in NOD/ShiLtJ mice as an animal model of SS. Treatment with interleukin (IL)-21, a major cytokine secreted from Tfh cells, suppressed Pax3 and Id3 expression via STAT3 in splenic T cells in vitro. Administration of pCMV14-3xFlag PAX3 vector improved the severity of SS by reducing the number of Tfh cells in NOD/ShiLtJ mice. Application of IL-21R-Fc increased the number of Pax3- and Id3-positive cells in the salivary glands, while reducing the proportion of Tfh cells and IL-17-producing T cells in NOD/ShiLtJ mice. The salivary glands from SS patients showed decreased levels of Pax3 or Id3 expression compared with healthy controls. Our findings regarding reinforcement of the Pax3-Id3 signal pathway may facilitate the development of novel therapeutic strategies for SS.


Subject(s)
Inhibitor of Differentiation Proteins/metabolism , Interleukins/pharmacology , Neoplasm Proteins/metabolism , PAX3 Transcription Factor/metabolism , Sjogren's Syndrome/immunology , T Follicular Helper Cells/immunology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Immunohistochemistry , Inhibitor of Differentiation Proteins/antagonists & inhibitors , Interleukin-17/metabolism , Interleukins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Neoplasm Proteins/antagonists & inhibitors , PAX3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Salivary Glands/drug effects , Salivary Glands/metabolism , Signal Transduction/drug effects , Sjogren's Syndrome/genetics , Sjogren's Syndrome/metabolism , Sjogren's Syndrome/therapy , Spleen/cytology , Spleen/drug effects , Spleen/metabolism , Up-Regulation
2.
World Neurosurg ; 131: 252-263.e2, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31376551

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is the most common and deadly form of brain tumor. After standard treatment of resection, radiotherapy, and chemotherapy, the 5-year survival is <5%. In recent years, research has uncovered several potential targets within the Notch signaling pathway, which may lead to improved patient outcomes. METHODS: A literature search was performed for articles containing the terms "Glioblastoma" and "Receptors, Notch" between 2003 and July 2015. Of the 62 articles retrieved, 46 met our criteria and were included in our review. Nine articles were identified from other sources and were subsequently included, leaving 55 articles reviewed. RESULTS: Of the 55 articles reviewed, 47 used established human GBM cell lines. Seventeen articles used human GBM surgical samples. Forty-five of 48 articles that assessed Notch activity showed increased expression in GBM cell lines. Targeting the Notch pathway was carried out through Notch knockdown and overexpression and targeting δ-like ligand, Jagged, γ-secretase, ADAM10, ADAM17, and Mastermindlike protein 1. Arsenic trioxide, microRNAs, and several other compounds were shown to have an effect on the Notch pathway in GBM. Notch activity in GBM was also shown to be associated with hypoxia and certain cancer-related molecular pathways such as PI3K/AKT/mTOR and ERK/MAPK. Most articles concluded that Notch activity amplifies malignant characteristics in GBM and targeting this pathway can bring about amelioration of these effects. CONCLUSIONS: Recent literature suggests targeting the Notch pathway has great potential for future therapies for GBM.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Neoplasm Proteins/antagonists & inhibitors , Receptors, Notch/antagonists & inhibitors , Signal Transduction/drug effects , ADAM Proteins/antagonists & inhibitors , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Arsenic Trioxide/pharmacology , Brain Neoplasms/blood supply , Cell Hypoxia , Cell Line, Tumor , Gene Knockdown Techniques , Glioblastoma/blood supply , Humans , Inhibitor of Differentiation Proteins/antagonists & inhibitors , Kruppel-Like Transcription Factors/antagonists & inhibitors , MicroRNAs/pharmacology , Microvessels , Molecular Targeted Therapy/methods , Netrin-1/antagonists & inhibitors , Niclosamide/pharmacology , Receptors, Notch/genetics , Receptors, Urokinase Plasminogen Activator/antagonists & inhibitors , Resveratrol/pharmacology , Signal Transduction/genetics , Tretinoin/pharmacology
3.
Cancer Gene Ther ; 22(9): 431-7, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26384138

ABSTRACT

The inhibitor of DNA-binding/differentiation 3 (Id3) protein is a helix-loop-helix transcription factor and may have an important role in cell proliferation and differentiation. This study was to evaluate the effects of upregulation of Id3 in human lung adenocarcinoma cells on proliferation, apoptosis, mobility and tumorigenicity. Short interference RNA suppression of Id3 (miRId3) in A549 cells was used to investigate the functional role(s) of Id3. Next, we used in vitro wound-healing assay and trans-well assay to study the effects of overexpressed Id3 on migration and invasion of A549 cells. Furthermore, to explore the influence of overexpressed Id3 on in vivo tumorigenesis, adenoviruses containing Id3 gene (Ad-Id3) and empty vector (Ad-LacZ) were generated. Co-transfection of pcDNA/miRId3 and pEGFP/Id3 into A549 cells reversed the Id3-induced cell proliferation inhibition and apoptosis. Upon Id3 transfection, A549 cells displayed decreased migratory and invasive capabilities, however, co-transfection of miRId3 and Id3 into A549 cells reversed the Id3-induced inhibitions of migratory and invasive capabilities. Three groups of nude mice were inoculated with Ad-LacZ, Ad-Id3 transfectants and untransfected A549 cells, respectively. Twenty-eight days after inoculation, tumors induced by Ad-Id3 transfectants grew much more slowly compared with Ad-LacZ transfectants and control group. This study provides for the first time both in vitro and in vivo proofs that forced expression of Id3 in lung adenocarcinoma cells reduces tumor growth rate and may be a potential target for tumor suppression.


Subject(s)
Adenocarcinoma/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Inhibitor of Differentiation Proteins/physiology , Lung Neoplasms/pathology , Neoplasm Proteins/physiology , Adenocarcinoma/genetics , Adenoviridae/genetics , Animals , Apoptosis , Carcinoma, Non-Small-Cell Lung/genetics , Cell Division , Cell Line, Tumor , Cell Movement , Female , Genetic Vectors , Humans , Inhibitor of Differentiation Proteins/antagonists & inhibitors , Inhibitor of Differentiation Proteins/biosynthesis , Inhibitor of Differentiation Proteins/genetics , Lung Neoplasms/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Transplantation , RNA Interference , RNA, Small Interfering/genetics , Transfection , Up-Regulation , Wound Healing
4.
Blood ; 126(5): 640-50, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26084673

ABSTRACT

Transcriptional regulators are recurrently altered through translocations, deletions, or aberrant expression in acute myeloid leukemia (AML). Although critically important in leukemogenesis, the underlying pathogenetic mechanisms they trigger remain largely unknown. Here, we identified that Id1 (inhibitor of DNA binding 1) plays a pivotal role in acute myeloid leukemogenesis. Using genetically modified mice, we found that loss of Id1 inhibited t(8;21) leukemia initiation and progression in vivo by abrogating protein kinase B (AKT)1 activation, and that Id1 interacted with AKT1 through its C terminus. An Id1 inhibitor impaired the in vitro growth of AML cells and, when combined with an AKT inhibitor, triggered even greater apoptosis and growth inhibition, whereas normal hematopoietic stem/progenitor cells were largely spared. We then performed in vivo experiments and found that the Id1 inhibitor significantly prolonged the survival of t(8;21)(+) leukemic mice, whereas overexpression of activated AKT1 promoted leukemogenesis. Thus, our results establish Id1/Akt1 signaling as a potential therapeutic target in t(8;21) leukemia.


Subject(s)
Inhibitor of Differentiation Protein 1/metabolism , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis , Carcinogenesis , Cell Line, Tumor , Disease Progression , Gene Knockdown Techniques , Humans , Inhibitor of Differentiation Protein 1/deficiency , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Proteins/antagonists & inhibitors , Inhibitor of Differentiation Proteins/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Interaction Domains and Motifs , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction , Translocation, Genetic
5.
Biochimie ; 112: 139-50, 2015 May.
Article in English | MEDLINE | ID: mdl-25778840

ABSTRACT

The four known ID proteins (ID1-4, Inhibitor of Differentiation) share a homologous helix loop helix (HLH) domain and act as dominant negative regulators of basic-HLH transcription factors. ID proteins also interact with many non-bHLH proteins in complex networks. The expression of ID proteins is increasingly observed in many cancers. Whereas ID-1, ID-2 and ID-3, are generally considered as tumor promoters, ID4 on the contrary has emerged as a tumor suppressor. In this study we demonstrate that ID4 heterodimerizes with ID-1, -2 and -3 and promote bHLH DNA binding, essentially acting as an inhibitor of inhibitors of differentiation proteins. Interaction of ID4 was observed with ID1, ID2 and ID3 that was dependent on intact HLH domain of ID4. Interaction with bHLH protein E47 required almost 3 fold higher concentration of ID4 as compared to ID1. Furthermore, inhibition of E47 DNA binding by ID1 was restored by ID4 in an EMSA binding assay. ID4 and ID1 were also colocalized in prostate cancer cell line LNCaP. The alpha helix forming alanine stretch N-terminal, unique to HLH ID4 domain was required for optimum interaction. Ectopic expression of ID4 in DU145 prostate cancer line promoted E47 dependent expression of CDKNI p21. Thus counteracting the biological activities of ID-1, -2 and -3 by forming inactive heterodimers appears to be a novel mechanism of action of ID4. These results could have far reaching consequences in developing strategies to target ID proteins for cancer therapy and understanding biologically relevant ID-interactions.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Inhibitor of Differentiation Protein 1 , Inhibitor of Differentiation Protein 2 , Inhibitor of Differentiation Proteins , Neoplasm Proteins , Prostatic Neoplasms , Transcription, Genetic , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Humans , Inhibitor of Differentiation Protein 1/antagonists & inhibitors , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Inhibitor of Differentiation Protein 2/antagonists & inhibitors , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/metabolism , Inhibitor of Differentiation Proteins/antagonists & inhibitors , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/metabolism , Male , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
6.
Br J Cancer ; 103(8): 1237-44, 2010 Oct 12.
Article in English | MEDLINE | ID: mdl-20842131

ABSTRACT

BACKGROUND: Inhibitors of DNA-binding proteins (Id1-4), lacking the basic DNA-binding domain, function as dominant inhibitors of cell-cycle regulators. Overexpression of Id proteins promotes cancer cell proliferation and resistance against apoptosis. Level of Id protein expression, especially of Id1, correlates with poor differentiation, enhanced malignant potential and more aggressive clinical behaviour of ovarian tumours. Although overexpression of Ids has been found and shown to correlate with poor clinical outcome, their inhibition at protein level has never been studied. METHODS: A peptide aptamer, Id1/3-PA7, targeting Id1 and Id3, was isolated from a randomised combinatorial expression library using yeast and mammalian two-hybrid systems. Id1/3-PA7 was fused, expressed and purified with a cell-penetrating protein transduction domain. RESULTS: Intracellular-delivered Id1/3-PA7 colocalised to Id1 and Id3. It induced cell-cycle arrest and apoptosis in ovarian cancer cells ES-2 and PA-1. It activated the E-box promoter and increased the expression level of cyclin-dependent kinase inhibitor (CDKN2A) in a dose-dependent manner that is paralleled by the cleavage of poly-ADP ribose polymerase. These effects were counteracted by ectopically overexpressed Id1 and Id3. CONCLUSION: Id1/3-PA7 could represent an exogenous anti-tumour agent that can significantly trigger cell-cycle arrest and apoptosis in ovarian cancer.


Subject(s)
Apoptosis/drug effects , Aptamers, Peptide/pharmacology , Carcinoma/pathology , Cell Cycle/drug effects , Inhibitor of Differentiation Proteins/antagonists & inhibitors , Ovarian Neoplasms/pathology , Antineoplastic Agents/pharmacology , Aptamers, Peptide/metabolism , Aptamers, Peptide/pharmacokinetics , Carcinoma/genetics , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Gene Expression Regulation, Neoplastic/drug effects , Genes, p16/drug effects , Humans , Inhibitor of Differentiation Protein 1/antagonists & inhibitors , Inhibitor of Differentiation Protein 1/metabolism , Inhibitor of Differentiation Proteins/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Ovarian Neoplasms/genetics , Tissue Distribution , Tumor Cells, Cultured
7.
Int J Oncol ; 37(3): 595-603, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20664928

ABSTRACT

Id3 is over-expressed in small cell lung cancer (SCLC). To test whether the tumourigenicity of SCLC cells can be inhibited by suppressing Id3 expression, we transfected siRNA into SCLC cell line GLC-19 and established two sublines (G-Id3-1 and G-Id3-7) which expressed only 30% of the level of Id3 measured in control transfectants. Suppression of Id3 expression in both G-Id3-1 and G-Id3-7 cells produced significant reductions in proliferation rates and in numbers of colonies formed in soft agar assay. When G-Id3-1, G-Id3-7 and the control transfectants were inoculated subcutaneously into 3 groups (8 each) of nude mice, respectively, all (100%) inoculated animals produced tumours. Although there was no difference in tumour incidents amongst the 3 groups, significant reductions were observed in both size and weight of tumours produced by either G-Id3-1 or G-Id3-7 cells. While the final average volume of tumours produced in control group was 1012.1+/-394 mm(3), it was significantly reduced (p<0.001, p<0.01) by 2.1- and 2.9-fold to 475.7+/-167 mm(3) and 354.3+/-218 mm(3) in groups inoculated with G-Id3-1 and G-Id3-7 cells, respectively. Similar differences were also observed in average weight of tumours. Upon induction of apoptosis by cytotoxin camptothecin, the percentages of apoptotic cells in G-Id3-1 and G-Id3-7 were, respectively >2.4-fold higher than that in control. The results in this study suggest that highly expressed Id3 in SCLC cells may be an important therapeutic target for tumour suppression.


Subject(s)
Inhibitor of Differentiation Proteins/biosynthesis , Lung Neoplasms/pathology , Neoplasm Proteins/biosynthesis , Small Cell Lung Carcinoma/pathology , Animals , Base Sequence , Camptothecin/pharmacology , Cell Growth Processes/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Inhibitor of Differentiation Proteins/antagonists & inhibitors , Inhibitor of Differentiation Proteins/genetics , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/therapy , Mice , Mice, Nude , Molecular Sequence Data , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/therapy , Transfection
8.
Breast Cancer Res Treat ; 124(3): 623-33, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20191379

ABSTRACT

Inhibitors of differentiation or DNA binding (Id) proteins have been shown to be involved in tumor growth, invasiveness, metastasis, and angiogenesis. Overexpression of Id proteins, especially Id1, correlates with unfavorable clinical prognosis. Thus, they are attractive molecular targets for anticancer therapy. Overexpression of Id proteins mediates breast cancer metastasis to lung. Targeting Id1 and Id3 expression in breast cancer cells reduces breast cancer metastasis in animal models. Different breast tumors failed to grow and/or metastasize in Id1 (+/-) Id3 (-/-) mice. Id1 and Id3 preferentially dimerize with the key regulatory E-proteins which inhibit the expression of different tumor suppressor genes. Nevertheless, the inhibition of tumorigenic activities of Id1 and Id3 at protein level has never been studied. Here, we isolated a novel peptide aptamer, Id1/3-PA7, specifically interacting with Id1 and Id3 from randomized combinatorial expression library using yeast and mammalian two-hybrid systems. Intracellular delivered Id1/3-PA7 co-localized to Id1 and Id3 and interfered with their functions. It repressed E47 protein sequestration by Id1 and Id3, activated the E-box promoter and increased the expression level of cyclin-dependent kinase inhibitors (CDKN1A and CDKN1B) in a dose-dependent fashion, paralleled by the cleavage of poly ADP ribose polymerase (PARP). These effects were counteracted by ectopically overexpressed Id1 and Id3. Peptide aptamer Id1/3-PA7 induced cell cycle arrest and apoptosis in breast cancer cells MCF7 and MDA-MB-231. In conclusion, Id1/3-PA7 could represent a nontoxic exogenous agent that can significantly provoke antiproliferative and apoptotic effects in breast cancer cells, which are associated with deregulated expression of Id1 and Id3.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Aptamers, Peptide/pharmacology , Breast Neoplasms/metabolism , Cell Cycle/drug effects , E-Box Elements/drug effects , Inhibitor of Differentiation Protein 1/antagonists & inhibitors , Inhibitor of Differentiation Proteins/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Promoter Regions, Genetic/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27 , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Neoplastic/drug effects , HeLa Cells , Humans , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Protein Binding , Transcription Factor 3/metabolism , Transfection , Two-Hybrid System Techniques , Up-Regulation
9.
Int J Oncol ; 33(1): 185-93, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18575765

ABSTRACT

Cdc42, a member of Rho GTPases family, is involved in the regulation of several cellular functions, such as rearrangement of actin cytoskeleton, membrane trafficking, cell-cycle progression, and transcriptional regulation. Aberrant expression or activity of Cdc42 has been reported in several tumours. Here, the specific role of Cdc42 in development and progression of colorectal cancer was analyzed through microarrays technology. A comparative analysis of Cdc42 overexpressing cells versus cells with decreased Cdc42 levels through siRNA revealed that Cdc42 overexpression down-regulated the potential tumour suppressor gene ID4. Results were validated by quantitative RT-PCR and the methylation status of the specific promoter, analyzed. Methylation-specific PCR and bisulfite sequencing PCR analysis revealed that Cdc42 induced the methylation of the CpG island of the ID4 promoter. Colorectal adenocarcinoma samples were compared with the corresponding adjacent normal tissue of the same patient in order to determine specific gene expression levels. The downregulation of ID4 by Cdc42 was also found of relevance in colorectal adenocarcinoma biopsies. Cdc42 was found to be overexpressed with high incidence (60%) in colorectal cancer samples, and this expression was associated with silencing of ID4 with statistical significance (p<0.05). Cdc42 may have a role in the development of colon cancer. Furthermore, inhibition of Cdc42 activity may have a direct impact in the management of colorectal cancer.


Subject(s)
Adenocarcinoma/chemistry , Colorectal Neoplasms/chemistry , Inhibitor of Differentiation Proteins/genetics , cdc42 GTP-Binding Protein/physiology , Adenocarcinoma/therapy , Aged , Aged, 80 and over , Colorectal Neoplasms/etiology , Colorectal Neoplasms/therapy , DNA Methylation , Down-Regulation , Epigenesis, Genetic , Female , Gene Silencing , Humans , Inhibitor of Differentiation Proteins/antagonists & inhibitors , Male , Middle Aged , Promoter Regions, Genetic , cdc42 GTP-Binding Protein/analysis , cdc42 GTP-Binding Protein/antagonists & inhibitors
10.
Trends Mol Med ; 12(12): 588-94, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17071138

ABSTRACT

In eukaryotic organisms, ID proteins are key regulators of development when they function to preserve the stem cell state and prevent lineage determination. By fueling several key features of tumor progression (deregulated proliferation, invasiveness, angiogenesis and metastasis), ID proteins contribute to multiple steps of tumorigenesis. Through oncogenic processes that lead to their aberrant activation in tumors, ID proteins transfer the phenotypic traits of embryonic stem cells to cancer cells. However, ID proteins have recently emerged as highly specialized factors in post-mitotic neurons. The elevated expression of ID proteins arrests neurons in the axon growth mode and prevents cessation of axonal elongation. Here, we discuss how unique properties of ID proteins in cancer cells and neurons pave the way to unexpected therapeutic opportunities.


Subject(s)
Inhibitor of Differentiation Proteins/physiology , Neoplasms/physiopathology , Animals , Cell Differentiation , Cell Proliferation , Humans , Inhibitor of Differentiation Proteins/antagonists & inhibitors , Inhibitor of Differentiation Proteins/genetics , Models, Biological , Models, Neurological , Neoplasms/drug therapy , Neoplasms/etiology , Neoplasms/pathology , Neurobiology , Neurons/cytology , Neurons/physiology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...