Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Cell Signal ; 46: 83-91, 2018 06.
Article in English | MEDLINE | ID: mdl-29501727

ABSTRACT

Neuroendocrine (NE) lung tumors account for 20% of total lung cancer cases and represent a subset of aggressive tumors with metastatic potential. High-risk NE lung cancer patients display disseminated disease, N-myc expression/amplification, and poorly differentiated tumors. In this study, we investigate the molecular mechanisms underlying a zinc-finger transcription factor, INSM1 in NE lung cancer. Our study revealed that INSM1 crosstalk with the Shh-PI3K/AKT-N-myc/Ascl1-MEK/ERK1/2 transcriptional network in NE lung cancer. The INSM1 expression pattern and functional data demonstrated that INSM1 is not only critical for NE differentiation, but also served as a NE tumor-specific marker in small cell lung carcinoma (SCLC). The Shh signaling pathway activates INSM1 expression through N-myc and Ascl1 in aggressive SCLC. The E2-box in the INSM1 promoter is the direct target recognized by N-myc and Ascl1 transcription factors. N-myc or Ascl1 activates endogenous INSM1 expression in lung cancer cells. INSM1 functions as a key player in NE lung cancer via Shh signaling that crosstalk with PI3K/AKT and MEK/ERK1/2 pathway to enhance N-myc stability in NE lung cancer. We investigate the negative effects of Shh inhibitor and knockdown of INSM1 in NE lung cancer cells. The combination of different Shh signaling pathway inhibitors targeting INSM1 and N-myc inhibits lung cancer cell growth and could be used as a new treatment option for SCLC.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hedgehog Proteins/metabolism , N-Myc Proto-Oncogene Protein/metabolism , Neuroendocrine Tumors/metabolism , Repressor Proteins/metabolism , Small Cell Lung Carcinoma/metabolism , Zinc Finger E-box Binding Homeobox 2/metabolism , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Promoter Regions, Genetic/genetics , Repressor Proteins/genetics , Signal Transduction
2.
J Biol Chem ; 291(21): 11313-22, 2016 May 20.
Article in English | MEDLINE | ID: mdl-27048654

ABSTRACT

INS-VNTR (insulin-variable number of tandem repeats) and AIRE (autoimmune regulator) have been associated with the modulation of insulin gene expression in thymus, which is essential to induce either insulin tolerance or the development of insulin autoimmunity and type 1 diabetes. We sought to analyze whether each functional domain of AIRE is critical for the activation of INS-VNTR in human thymic epithelial cells. Twelve missense or nonsense mutations in AIRE and two chimeric AIRE constructs were generated. A luciferase reporter assay and a pulldown assay using biotinylated INS-class I VNTR probe were performed to examine the transactivation and binding activities of WT, mutant, and chimeric AIREs on the INS-VNTR promoter. Confocal microscopy analysis was performed for WT or mutant AIRE cellular localization. We found that all of the AIRE mutations resulted in loss of transcriptional activation of INS-VNTR except mutant P252L. Using WT/mutant AIRE heterozygous forms to modulate the INS-VNTR target revealed five mutations (R257X, G228W, C311fsX376, L397fsX478, and R433fsX502) that functioned in a dominant negative fashion. The LXXLL-3 motif is identified for the first time to be essential for DNA binding to INS-VNTR, whereas the intact PHD1, PHD2, LXXLL-3, and LXXLL-4 motifs were important for successful transcriptional activation. AIRE nuclear localization in the human thymic epithelial cell line was disrupted by mutations in the homogenously staining region domain and the R257X mutation in the PHD1 domain. This study supports the notion that AIRE mutation could specifically affect human insulin gene expression in thymic epithelial cells through INS-VNTR and subsequently induce either insulin tolerance or autoimmunity.


Subject(s)
Epithelial Cells/metabolism , Thymus Gland/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Cell Line , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Epithelial Cells/pathology , Humans , Mutation , Protein Structure, Tertiary , Thymus Gland/pathology , Transcription Factors/genetics , AIRE Protein
3.
BMC Pulm Med ; 16: 49, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27072116

ABSTRACT

BACKGROUND: Insulinoma associated-1 (INSM1) gene is expressed exclusively in early embryonic neuroendocrine tissues, but has been found highly re-activated in most of the neuroendocrine tumors including small cell lung carcinoma. METHODS: In order to elucidate the functional effects of INSM1 in normal lung development, we used a conditional lung-specific INSM1 transgenic mouse model. Transgenic (Tet-on system) CMV-INSM1 responder mice were bred with the lung-specific, club cell secretory protein (CCSP) promoter-rtTA activator mice to produce bi-transgenic progeny carrying both alleles, CCSP-rtTA and Tet-on-INSM1. Mice were fed with doxycycline containing food at the initial mating day to the postnatal day 21. Lung samples were collected at embryonic day 17.5, newborn, and postnatal day 21 for analyses. RESULTS: Northern blot, RT-PCR, and immunohistochemical analyses revealed that doxycycline induced respiratory epithelium-specific INSM1 expression in bi-transgenic mice. Samples from postnatal day 21 mice revealed a larger lung size in the bi-transgenic mouse as compared to the single-transgenic or wild-type littermates. The histopathology results showed that the alveolar space in the bi-transgenic mice were 4 times larger than those in the single transgenic or wild-type littermates. In contrast, the size was not significantly different in the lungs collected at E17.5 or newborn among the bi-transgenic, single transgenic, or wild type mice. The respiratory epithelium with INSM1 ectopic expression suppressed cyclin D1 signal. Further in vitro studies revealed that the ectopic expression of INSM1 suppresses cyclin D1 expression and delays cell cycle progression. CONCLUSION: The current study suggests that CCSP promoter-driven INSM1 ectopic expression impairs normal lung development especially in postnatal alveologenesis.


Subject(s)
DNA-Binding Proteins/genetics , Ectopic Gene Expression/genetics , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Lung Neoplasms/metabolism , Pulmonary Alveoli/embryology , Small Cell Lung Carcinoma/metabolism , Transcription Factors/genetics , Animals , Blotting, Northern , Blotting, Western , Bronchi/cytology , Bronchi/metabolism , Case-Control Studies , Cell Line , Cyclin D1/metabolism , DNA-Binding Proteins/metabolism , Epithelial Cells/cytology , Flow Cytometry , Humans , Immunohistochemistry , Lung/embryology , Lung/growth & development , Lung/metabolism , Mice , Mice, Transgenic , Pulmonary Alveoli/growth & development , Pulmonary Alveoli/metabolism , Repressor Proteins , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism
4.
Cell Oncol (Dordr) ; 39(3): 279-86, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26902080

ABSTRACT

BACKGROUND: Insulinomas are the most common type of neuroendocrine (NE) pancreatic islet tumors. Patients with insulinomas may develop complications associated with hyperinsulinemia. To increase the treatment options for insulinoma patients, we have tested a conditionally replicating adenovirus that has been engineered in such a way that it can specifically express therapeutic genes in NE tumors. METHODS: We used a promoter-specific adenoviral vector delivery system that is regulated by an INSM1 (insulinoma-associated-1) promoter, which is silent in normal adult tissues but active in developing NE cells and tumors. Through a series of modifications, using an insulator (HS4) and neuron-restrictive silencer elements (NRSEs), an oncolytic adenoviral vector was generated that retains tumor specificity and drives the expression of a mutated adenovirus E1A gene (Δ24E1A) and the herpes simplex virus thymidine kinase (HSV-tk) gene. The efficacy of this vector was tested in insulinoma-derived MIN, RIN, ßTC-1 and pancreatic (Panc-1) cells using in vitro cell survival and in vivo tumor growth assays. RESULTS: Using in vitro insulinoma-derived cell lines and an in vivo subcutaneous mouse tumor model we found that the INSM1 promoter-driven viruses were able to replicate specifically in INSM1-positive cells. INSM1-specific HSV-tk expression in combination with ganciclovir treatment resulted in dose-dependent tumor cell killing, leaving INSM1-negative cells unharmed. When we combined the INSM1-promoter driven HSV-tk with Δ24E1A and INSM1p-HSV-tk (K5) viruses, we found that the co-infected insulinoma-derived cells expressed higher levels of HSV-tk and exhibited more efficient tumor suppression than cells infected with INSM1p-HSV-tk virus alone. CONCLUSIONS: INSM1 promoter-driven conditionally replicating adenoviruses may serve as a new tool for the treatment of insulinoma and may provide clinicians with additional options to combat this disease.


Subject(s)
Genetic Therapy/methods , Insulinoma , Oncolytic Virotherapy/methods , Pancreatic Neoplasms , Promoter Regions, Genetic , Repressor Proteins/genetics , Adenoviridae , Animals , Antineoplastic Agents/pharmacology , Blotting, Western , Cell Line, Tumor , Ganciclovir/pharmacology , Genetic Vectors , Humans , Mice , Thymidine Kinase/genetics , Xenograft Model Antitumor Assays
5.
Int J Oncol ; 48(1): 173-80, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26530405

ABSTRACT

Accurate detection of neuroendocrine (NE) tumors is critically important for better prognosis and treatment outcomes in patients. To demonstrate the efficacy of using an adenoviral vector for the detection of NE tumors, we have constructed a pair of adenoviral vectors which, in combination, can conditionally replicate and release Gaussia luciferase into the circulation after infecting the NE tumors. The expression of these two vectors is regulated upstream by an INSM1-promoter (insulinoma-associated-1) that is specifically active in NE tumors and developing NE tissues, but silenced in normal adult tissues. In order to retain the tumor-specificity of the INSM1 promoter, we have modified the promoter using the core insulator sequence from the chicken ß-globin HS4 insulator and the neuronal restrictive silencing element (NRSE). This modified INSM1-promoter can retain NE tumor specificity in an adenoviral construct while driving a mutated adenovirus E1A gene (∆24E1A), the Metridia, or Gaussia luciferase gene. The in vitro cell line and mouse xenograft human tumor studies revealed the NE specificity of the INSM1-promoter in NE lung cancer, neuroblastoma, medulloblastoma, retinoblastoma, and insulinoma. When we combined the INSM1-promoter driven Gaussia luciferase with ∆24E1A, the co-infected NE tumor secreted higher levels of Gaussia luciferase as compared to the INSM1p-Gaussia virus alone. In a mouse subcutaneous xenograft tumor model, the combination viruses secreted detectable level of Gaussia luciferase after infecting an INSM1-positive NE lung tumor for ≥12 days. Therefore, the INSM1-promoter specific conditional replicating adenovirus represents a sensitive diagnostic tool to aid clinicians in the detection of NE tumors.


Subject(s)
Genetic Therapy , Luciferases/genetics , Neuroendocrine Tumors/genetics , Repressor Proteins/genetics , Adenoviridae/genetics , Animals , Carcinoma, Neuroendocrine , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Genetic Vectors , Humans , Luciferases/biosynthesis , Mice , Neuroendocrine Tumors/diagnosis , Neuroendocrine Tumors/pathology , Promoter Regions, Genetic
6.
Oncotarget ; 6(34): 36700-12, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26456864

ABSTRACT

Insulinoma associated-1 (IA-1/INSM1) gene is exclusively expressed during early embryonic development, but has been found to be re-expressed at high levels in neuroendocrine tumors including neuroblastoma. Using over-expression and knockdown experiments in neuroblastoma cells, we showed that INSM1 is critical for cell proliferation, BME-coated invasion, and soft agar colony formation. Here, we identified INSM1 as a novel target gene activated by N-myc in N-myc amplified neuroblastoma cells. The Sonic hedgehog signaling pathway induced INSM1 by increasing N-myc expression. INSM1 activated PI3K/AKT/GSK3ß pathways to suppress N-myc phosphorylation (Thr-58) and inhibited degradation of N-myc. Inversely, N-myc protein bound to the E2-box region of the INSM1 promoter and activated INSM1 expression. The invasion assay and the xenograft nude mouse tumor model revealed that the INSM1 factor facilitated growth and oncogenesis of neuroblastoma. The current data supports our hypothesis that a positive-feedback loop of sonic hedgehog signaling induced INSM1 through N-myc and INSM1 enhanced N-myc stability contributing to the transformation of human neuroblastoma.


Subject(s)
N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/metabolism , Repressor Proteins/metabolism , Animals , Carcinogenesis , Cell Line, Tumor , Cell Proliferation/physiology , Glycogen Synthase Kinase 3 beta/metabolism , Heterografts , Humans , Mice , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Neuroblastoma/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt/metabolism , Repressor Proteins/genetics , Signal Transduction , Transcription Factors/metabolism
7.
Life Sci ; 114(1): 45-50, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25128855

ABSTRACT

AIMS: Differentiation of pancreatic endocrine cells depends upon the activation of genes that are regulated by islet transcription factors (ITFs). Evidence suggests that ITFs contribute to the development of the pancreas. These studies are focused on developing a system to deliver individual ITF from different stages of islet cell development to promote precursors or other cell types to trans-differentiate into islet-like insulin-positive cells. MAIN METHODS: Protein transduction domains (PTDs) derived from the HIV-TAT peptide (YGRKKRRQRRR) were fused with three ITFs, Ngn3, Pdx1, and NeuroD/ß2, to facilitate the uptake of ITF recombinant proteins into various cell types. The three TAT-fused ITFs, Ngn3, Pdx1, and NeuroD/ß2 were constructed in a bacterial 6×His affinity tag-TAT recombinant protein expression system. The recombinant proteins were expressed using IPTG induction and purified to homogeneity using a nickel affinity column. KEY FINDINGS: The biological activity of each TAT-fused ITF was demonstrated by nuclear translocation, induction of target gene promoter activity, and the trans-differentiation of pancreatic acinar cells, AR42J, into insulin-positive cells. SIGNIFICANCE: This study provides advanced information for developing strategies using recombinant TAT-fused ITF proteins in place of adenoviral vectors for the conversion of pancreatic exocrine cells into insulin-positive cells for the treatment of diabetes.


Subject(s)
Endocrine Cells/cytology , Gene Products, tat/genetics , Insulin/metabolism , Islets of Langerhans/cytology , Transcription Factors/genetics , Acinar Cells/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Cell Line , Chromatography, Affinity , Cricetinae , Genetic Vectors , Homeodomain Proteins/genetics , Humans , Mice , Nerve Tissue Proteins/genetics , Promoter Regions, Genetic , Rats , Recombinant Proteins/genetics , Trans-Activators/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics
8.
Cell Signal ; 26(4): 740-7, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24407176

ABSTRACT

INSM1 is an islet transcription factor essential for pancreas development. INSM1 functions as a transcriptional repressor of NeuroD/ß2 and insulin gene in the pancreas. INSM1 also possesses extra-nuclear activities through binding to multiple cellular regulators such as cyclin D1 and RACK1. In this study, we report that the interaction of INSM1 and RACK1 is essential to enhance the insulin receptor (InR)-mediated signaling pathway. A proline-rich region in the N-terminus of INSM1 is required for RACK1 binding, which interrupts RACK1-InR interaction and enhances InR signal activation. Binding of INSM1 to RACK1 increases AKT phosphorylation. The INSM1-enhanced AKT phosphorylation can be inhibited by the PI3K inhibitor, LY294002. When INSM1 induces AR42J cell trans-differentiation, the Nkx6.1 gene is activated through the InR-mediated signaling pathway and an elevation of the acetyl-H4 modification on the Nkx6.1 gene promoter/enhancer is observed. The PI3K inhibitor interrupts Nkx6.1 and insulin gene expression. Therefore, we conclude that the extra-nuclear activity of INSM1 by enhancing the PI3K/AKT signaling pathway is important for pancreatic cell differentiation.


Subject(s)
DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Neuropeptides/metabolism , Signal Transduction , Transcription Factors/metabolism , Amino Acid Sequence , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Chromones/pharmacology , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Homeodomain Proteins/genetics , Insulin/genetics , Insulin/metabolism , Mice , Molecular Sequence Data , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptor, Insulin/metabolism , Receptors for Activated C Kinase , Repressor Proteins , Transcription Factors/chemistry , Transcription Factors/genetics
9.
Virol J ; 10: 86, 2013 Mar 14.
Article in English | MEDLINE | ID: mdl-23497017

ABSTRACT

BACKGROUND: Lung cancer is the leading cause of cancer-related deaths in the US. Recombinant vectors based on adeno-associated virus (AAV) and lentivirus are promising delivery tools for gene therapy due to low toxicity and long term expression. The efficiency of the gene delivery system is one of the most important factors directly related to the success of gene therapy. METHODS: We infected SCLC cell lines, SHP-77, DMS 53, NCI-H82, NCI-H69, NCI-H727, NCI-H1155, and NSCLC cell lines, NCI-H23, NCI-H661, and NCI-H460 with VSV-G pseudo-typed lentivirus or 5 AAV serotypes, AAV2/1, AAV2/2, AAV2/4, AAV2/5, and AAV2/8 expressing the CMV promoter mCherry or green fluorescent protein transgene (EGFP). The transduction efficiency was analyzed by fluorescent microscopy and flow cytometry. RESULTS: Of all the serotypes of AAV examined, AAV2/1 was the optimal serotype in most of the lung cancer cell lines except for NCI-H69 and NCI-H82. The highest transduction rate achieved with AAV2/1 was between 30-50% at MOI 100. Compared to all AAV serotypes, lentivirus had the highest transduction efficiency of over 50% at MOI 1. Even in NCI-H69 cells resistant to all AAV serotypes, lentivirus had a 10-40% transduction rate. To date, AAV2 is the most widely-used serotype to deliver a transgene. Our results showed the transduction efficiency of AAVs tested was AAV2/1 > AA2/5 = AAV2/2> > AAV2/4 and AAV2/8. CONCLUSIONS: This study demonstrated that VSV-G pseudotyped lentivirus and AAV2/1 can mediate expression of a transgene for lung cancer gene therapy. Overall, our results showed that lentivirus is the best candidate to deliver a transgene into lung cancer cells for treatment.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Lentivirus/genetics , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Membrane Glycoproteins/genetics , Transduction, Genetic/methods , Viral Envelope Proteins/genetics , Cell Line, Tumor , Dependovirus/physiology , Genetic Therapy/instrumentation , Genetic Vectors/genetics , Genetic Vectors/physiology , Humans , Lentivirus/metabolism , Lung Neoplasms/virology , Membrane Glycoproteins/metabolism , Transduction, Genetic/instrumentation , Viral Envelope Proteins/metabolism
10.
Ochsner J ; 13(1): 91-9, 2013.
Article in English | MEDLINE | ID: mdl-23533081

ABSTRACT

BACKGROUND: Insulinoma-associated protein 1 (INSM1) is a zinc finger transcriptional repressor with a limited spatial and temporal embryonic expression pattern in neuronal and neuroendocrine tissues. Interestingly, INSM1 activity is reactivated in neuroendocrine tumors such as small-cell lung cancer (SCLC), neuroblastoma, medulloblastoma, and retinoblastoma. Adenoviral constructs with the 1.7-kilobase pair INSM1 promoter-driven herpes simplex virus thymidine kinase (HSV-tk) gene could effectively suppress D283 Med subcutaneous xenograft tumor growth. Undesirably, sequences in the adenoviral backbone overrode promoter specificity in vivo. Incorporation of both the chicken ß-globin HS4 insulator sequence and 2 copies of the mouse nicotinic acetylcholine receptor (nAchR) neuronal restrictive silencer element abolished the nonspecific activation of the INSM1 promoter in vivo. METHODS: The luciferase reporter gene was replaced with the HSV-tk suicide gene to generate the Ad-K5 virus. Both in vitro cell viability assays and in vivo tumor regression studies were used to determine the efficacy of the improved configuration INSM1 promoter adenoviral construct against a panel of neuroendocrine cell lines. RESULTS: In vitro cell viability assays with the Ad-K5 HSV-tk-expressing construct further reinforced that the Ad-K5 virus could eradicate SCLC, insulinoma, medulloblastoma, and neuroblastoma cells. Further, Ad-K5 virus treatment of a D283 Med subcutaneous xenograft tumor showed a superior antitumor effect over the control Ad-RSV (Rous sarcoma virus)-HSV-tk. CONCLUSIONS: Improvements to the INSM1 promoter resulted in a stronger and more selective adenovirus. Treatment of a panel of neuroendocrine carcinomas with the Ad-K5 virus revealed enhanced antitumor activity over the RSV control, demonstrating its usefulness for the treatment of a variety of neuroendocrine tumors.

11.
J Cell Physiol ; 227(6): 2470-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21830214

ABSTRACT

In this study, the functional role of INSM1 is examined with an AR42J acinar cell model for trans-differentiation into insulin-positive cells. Islet transcription factors (ITFs: INSM1, Pdx-1, and NeuroD1) are over-expressed in AR42J cells using adenoviral vectors. Addition of Ad-INSM1 alone or the combination of three ITFs to the AR42J cells triggers cellular trans-differentiation. Ectopic expression of INSM1 directly induces insulin, Pax6, and Nkx6.1 expression, whereas Pdx-1 and NeuroD1 were slightly suppressed by INSM1. Addition of Pdx-1 and NeuroD1 with INSM1 further enhances endocrine trans-differentiation by increasing both the numbers and intensity of the insulin-positive cells with simultaneous activation of ITFs, Ngn3 and MafA. INSM1 expression alone partially inhibits dexamethasone-induced exocrine amylase expression. The combination of the three ITFs completely inhibits amylase expression and concomitantly induces greater acinar cell trans-differentiation into endocrine cells. Also, addition of the three ITFs promotes EGF and TGFß receptors expression. Stimulation by the three ITFs along with the EGF/TGFß growth factors strongly promotes insulin gene expression. The combination of the three ITFs and EGF/TGFß growth factors with the primary cultured pancreatic acini also facilitates exocrine to endocrine cell differentiation. Taken together, both the AR42J cell line and the primary cultured mouse acinar cells support INSM1 induced acini trans-differentiation model.


Subject(s)
Acinar Cells/metabolism , Cell Transdifferentiation , Islets of Langerhans/metabolism , Repressor Proteins/metabolism , Acinar Cells/drug effects , Acinar Cells/pathology , Adenoviridae/genetics , Amylases/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Cell Transdifferentiation/drug effects , Cricetinae , Dexamethasone/pharmacology , ErbB Receptors/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Genetic Vectors , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Insulin/genetics , Insulin/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/pathology , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , RNA, Messenger/metabolism , Rats , Receptors, Transforming Growth Factor beta/metabolism , Repressor Proteins/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Transfection
12.
Diabetes ; 60(1): 336-44, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20876716

ABSTRACT

OBJECTIVE: Polymorphic INS-VNTR plays an important role in regulating insulin transcript expression in the human thymus that leads to either insulin autoimmunity or tolerance. The molecular mechanisms underlying the INS-VNTR haplotype-dependent insulin expression are still unclear. In this study, we determined the mechanistic components underlying the differential insulin gene expression in human thymic epithelial cells, which should have profound effects on the insulin autoimmune tolerance induction. RESEARCH DESIGN AND METHODS: A repetitive DNA region designated as a variable number of tandem repeats (VNTR) is located upstream of the human insulin gene and correlates with the incidence of type 1 diabetes. We generated six class I and two class III VNTR constructs linked to the human insulin basal promoter or SV40 heterologous promoter/enhancer and demonstrated that AIRE protein modulates the insulin promoter activities differentially through binding to the VNTR region. RESULTS: Here we show that in the presence of the autoimmune regulator (AIRE), the class III VNTR haplotype is responsible for an average of three-fold higher insulin expression than class I VNTR in thymic epithelial cells. In a protein-DNA pull-down experiment, AIRE protein is capable of binding to VNTR class I and III probes. Further, the transcriptional activation of the INS-VNTR by AIRE requires the insulin basal promoter. The VNTR sequence loses its activation activity when linked to a heterologous promoter and/or enhancer. CONCLUSIONS: These findings demonstrate a type 1 diabetes predisposition encoded by the INS-VNTR locus and a critical function played by AIRE, which constitute a dual control mechanisms regulating quantitative expression of insulin in human thymic epithelial cells.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Epithelial Cells/physiology , Insulin/genetics , Polymorphism, Genetic , Tandem Repeat Sequences/genetics , Thymus Gland/physiology , Autoimmunity/genetics , Cloning, Molecular , DNA Primers , Enhancer Elements, Genetic/genetics , Exons/genetics , Gene Expression Regulation , Genes, Reporter , Humans , Luciferases/genetics , Minisatellite Repeats/genetics , Polymerase Chain Reaction , Promoter Regions, Genetic/genetics , Restriction Mapping , Transcription, Genetic
13.
Endocrinology ; 151(5): 2030-9, 2010 May.
Article in English | MEDLINE | ID: mdl-20215568

ABSTRACT

Insulinoma-associated antigen-1 (INSM1/IA-1) is a unique zinc-finger transcription factor restrictedly expressed in pancreatic beta-cells during early pancreas development. INSM1 is transiently activated by the islet-specific endocrine factor neurogenin 3, and it subsequently regulates downstream target genes NeuroD1 and insulin during beta-cell maturation. Here, we examined how the INSM1 transcription factor contributes to endocrine cell differentiation using a defined serum-free medium-primed pancreatic duct cell model. We showed that ectopic expression of INSM1 can promote Panc-1 cell trans-differentiation. INSM1 up-regulates two islet transcription factors (ITFs), paired box 6 and homeodomain transcription factor 6.1, whereas other ITFs, including pancreatic duodenal homeobox-1 (Pdx-1), homeodomain transcription factor 2.2, NeuroD1, paired box 4, and neurogenin 3, were either down-regulated or absent. The result suggests that INSM1 is capable of regulating multiple ITFs and the insulin gene either directly or indirectly. When we overexpressed three ITFs, INSM1/Pdx-1/NeuroD1, in the Panc-1 differentiation model, higher insulin expression was observed in parallel with the activation of an additional ITF, neurogenin 3, signifying endocrine cell activation. Insulin expression from the three ITFs stimulation was readily detected by immunostaining and increased 40% as compared with the insulin-transferrin-selenium-LacZ control. Furthermore, we examined the differential chromatin acetylation patterns within the insulin promoter region using the chromatin immunoprecipitation assay. INSM1 alone can selectively enhance acetylation of histone H4, whereas NeuroD1 and Pdx-1 favor the acetylation of histone H3. Both H3 and H4 histone acetylations facilitate insulin gene expression. The consistent functional effect of INSM1, either with or without other ITFs, promotes pancreatic duct cell differentiation as well as induces Panc-1 cell cycle arrest.


Subject(s)
Cell Transdifferentiation , Repressor Proteins/physiology , Transcription Factors/physiology , Zinc Fingers , Acetylation , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Cell Cycle , Cell Line, Tumor , Eye Proteins/genetics , Eye Proteins/metabolism , Flow Cytometry , Histones/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Insulin/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Pancreatic Ducts/metabolism , Pancreatic Ducts/pathology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection
14.
Hum Gene Ther ; 20(11): 1308-18, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19604042

ABSTRACT

The INSM1 gene encodes a developmentally regulated zinc finger transcription factor. INSM1 expression is normally absent in adult tissues, but is reactivated in neuroendocrine tumor cells. In the present study, we analyzed the therapeutic potential of an adenoviral INSM1 promoter-driven herpes simplex virus thymidine kinase (HSV-tk) construct in primitive neuroectodermal tumors (PNETs). We constructed an adenoviral INSM1 promoter-driven HSV-tk gene for therapy in PNETs. The PNET-specific adeno-INSM1 promoter HSV-tk construct was tested both in vitro and in vivo in a nude mouse tumor model. Northern blot analysis and transient transfection of an INSM1 promoter-driven luciferase reporter gene indicated that the INSM1 promoter was active in neuroblastoma (IMR-32), retinoblastoma (Y79), and medulloblastoma (D283 Med) cells, but not in glioblastoma (U-87 MG) cells. After Ad-INSM1p-HSV-tk infection, the levels of HSV-tk protein expression were consistent with INSM1 promoter activities. Furthermore, in vitro multiplicity of infection and ganciclovir (GCV) sensitivity studies indicated that the INSM1 promoter could mediate specific expression of the HSV-tk gene and selective killing of INSM1-positive PNETs. In vivo intratumoral adenoviral delivery demonstrated that the INSM1 promoter could direct HSV-tk gene expression in a nude mouse tumor model and effectively repressed tumor growth in response to GCV treatment. Taken together, our data show that the INSM1 promoter is specific and effective for targeted cancer gene therapy in PNETs.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Genetic Therapy/methods , Genetic Vectors/immunology , Neuroectodermal Tumors, Primitive/therapy , Promoter Regions, Genetic/genetics , Repressor Proteins/genetics , Thymidine Kinase/genetics , Adenoviridae/genetics , Animals , Antiviral Agents/therapeutic use , Blotting, Northern , Blotting, Western , Cell Line, Tumor , Ganciclovir/therapeutic use , Genetic Vectors/genetics , Humans , Male , Mice , Mice, Nude , Neuroectodermal Tumors, Primitive/drug therapy , Neuroectodermal Tumors, Primitive/genetics , Tetrazolium Salts , Thiazoles , Viral Proteins/genetics , beta-Galactosidase
15.
FASEB J ; 23(7): 2024-33, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19246490

ABSTRACT

Zinc-finger transcription factors are DNA-binding proteins that are implicated in many diverse biological functions. INSM1 (formerly IA-1) contains five zinc-finger motifs and functions as a transcription factor. INSM1 protein structure is highly conserved in homologues of different species. It is predominantly expressed in developing neuroendocrine tissues and the nervous system in mammals. INSM1 represents an important player in early embryonic neurogenesis. In pancreatic endocrine cell differentiation, Ngn3 first activates INSM1 and subsequently NeuroD/beta2. Conversely, INSM1 exerts a feedback mechanism to suppress NeuroD/beta2 and its own gene expression. INSM1 gene ablation in the mouse results in the impairment of pancreatic endocrine cell maturation. Further, deletion of INSM1 severely impairs catecholamine biosynthesis and secretion from the adrenal gland that results in early embryonic lethality. Genetically, INSM1 acts as a downstream factor of Mash 1 and Phox2b in the differentiation of the sympatho-adrenal lineage. In the developing neocortex, mouse embryos lacking INSM1 expression contain half the number of basal progenitors and show a reduction in cortical plate radial thickness. Cell signaling studies reveal that INSM1 contributes to the induction of cell cycle arrest/exit necessary to facilitate cellular differentiation. INSM1 is highly expressed in tumors of neuroendocrine origin. Hence, its promoter could serve as a tumor-specific promoter that drives a specific targeted cancer gene therapy for the treatment of neuroendocrine tumors. Taken together, all of these features of INSM1 strongly support its role as an important regulator during neuroendocrine differentiation.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/physiology , Neuroendocrine Cells/cytology , Repressor Proteins/physiology , Carcinoma, Neuroendocrine/chemistry , Carcinoma, Neuroendocrine/pathology , DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , Humans , Pancreas/cytology , Repressor Proteins/analysis , Repressor Proteins/genetics , Transcription Factors/physiology
16.
J Biol Chem ; 284(9): 5574-81, 2009 Feb 27.
Article in English | MEDLINE | ID: mdl-19124461

ABSTRACT

INSM1 is a zinc finger transcription factor that plays an important role in pancreatic beta-cell development. To further evaluate its role in cell fate determination, we investigated INSM1 effects on cell cycle function. The cyclin box of cyclin D1 is essential for INSM1 binding. Competitive pull-down and co-immunoprecipitation revealed that INSM1 binding to cyclin D1 interrupts its association with CDK4 and induces hypophosphorylation of the retinoblastoma protein. An inducible Tet-on system was established in Cos-7 and Panc-1 cells. Using serum starvation, we synchronized the cell cycle and subsequently induced cell cycle progression by serum stimulation. Comparison of the INSM1 induction group with the noninduced control group, INSM1 ectopic expression causes cell cycle arrest, whereas the INSM1-mediated cell cycle arrest could be reversed by cyclin D1 and CDK4 overexpression. The proline-rich N-terminal portion of INSM1 is required for cyclin D1 binding. Mutation of proline residues abolished cyclin D1 binding and also diminished its ability to induce cell cycle arrest. Cellular proliferation of Panc-1 cells was inhibited by INSM1 overexpression demonstrated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, soft agar colony formation, as well as tumor growth in a nude mouse model. Taken together, we provide evidence to support that INSM1 binds to cyclin D1, interrupts cell cycle signaling, and inhibits cellular proliferation.


Subject(s)
Cell Cycle/physiology , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/metabolism , DNA-Binding Proteins/metabolism , Pancreatic Neoplasms/pathology , Repressor Proteins/metabolism , Zinc Fingers , Animals , COS Cells , Cell Proliferation , Cells, Cultured , Chlorocebus aethiops , Cyclin D1/genetics , Cyclin-Dependent Kinase 4/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Humans , Immunoprecipitation , Insulin/genetics , Insulin/metabolism , Pancreatic Neoplasms/metabolism , Phosphorylation , Promoter Regions, Genetic , Protein Interaction Domains and Motifs , RNA, Small Interfering/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Two-Hybrid System Techniques
17.
J Endocrinol ; 198(1): 29-39, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18417529

ABSTRACT

In this study, an insulinoma-associated antigen-1 (INSM1)-binding site in the proximal promoter sequence of the insulin gene was identified. The co-transfection of INSM1 with rat insulin I/II promoter-driven reporter genes exhibited a 40-50% inhibitory effect on the reporter activity. Mutational experiments were performed by introducing a substitution, GG to AT, into the INSM1 core binding site of the rat insulin I/II promoters. The mutated insulin promoter exhibited a three- to 20-fold increase in the promoter activity over the wild-type promoter in several insulinoma cell lines. Moreover, INSM1 overexpression exhibited no inhibitory effect on the mutated insulin promoter. Chromatin immunoprecipitation assays using beta TC-1, mouse fetal pancreas, and Ad-INSM1-transduced human islets demonstrated that INSM1 occupies the endogenous insulin promoter sequence containing the INSM1-binding site in vivo. The binding of the INSM1 to the insulin promoter could suppress approximately 50% of insulin message in human islets. The mechanism for transcriptional repression of the insulin gene by INSM1 is mediated through the recruitment of cyclin D1 and histone deacetylase-3 to the insulin promoter. Anti-INSM1 or anti-cyclin D1 morpholino treatment of fetal mouse pancreas enhances the insulin promoter activity. These data strongly support the view that INSM1 is a new zinc-finger transcription factor that modulates insulin gene transcription during early pancreas development.


Subject(s)
DNA-Binding Proteins/physiology , Insulin/genetics , Promoter Regions, Genetic , Repressor Proteins/physiology , Animals , Binding Sites , Cell Line, Tumor , Cyclin D1/physiology , Gene Expression Regulation , Histone Deacetylases/physiology , Humans , Mice , Pancreas/embryology , Rats , Transcription, Genetic
18.
J Pediatr Gastroenterol Nutr ; 45(4): 493-6, 2007 Oct.
Article in English | MEDLINE | ID: mdl-18030223

ABSTRACT

A vaccine could alleviate major morbidity and mortality associated with Helicobacter pylori infection. We immunized BALB/c mice with 3 doses of a protein or DNA vaccine based on H pylori urease B. Protein alone was immunogenic even after the first dose, whereas DNA did not elicit antibodies after 3 doses. DNA preceding protein (D-P-P) appeared to blunt the response to protein, whereas DNA following protein (P-D-D) shifted from a predominantly T helper 2 (Th2) profile to a balanced Th1:Th2 profile. These preliminary findings may have important implications for the development of an H pylori vaccine.


Subject(s)
Bacterial Proteins/therapeutic use , Bacterial Vaccines/therapeutic use , DNA/immunology , Helicobacter pylori/enzymology , Urease/immunology , Vaccines, DNA/therapeutic use , Animals , Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Female , Helicobacter Infections/prevention & control , Helicobacter pylori/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Vaccination/methods , Vaccines, DNA/immunology
19.
FEBS Lett ; 581(5): 949-54, 2007 Mar 06.
Article in English | MEDLINE | ID: mdl-17300785

ABSTRACT

INSM1 is a downstream target gene of neurogenin 3 (ngn3). A promoter construct containing the -426/+40bp region transiently co-transfected into NIH-3T3 cells with a ngn3 expression plasmid resulted in a 12-fold increase in promoter activity. The ngn3/E47 heterodimer selectively binds and activates the E-box3 of the INSM1 promoter. The endogenous ngn3 and CREB-binding protein (CBP) co-activator occupy the INSM1 promoter, resulting in hyper-acetylation of histone H3/H4 chromatin in a human neuroblastoma cell line, IMR-32. Additionally, adenoviral ngn3 can induce endogenous INSM-1 expression in pancreatic ductal carcinoma-1 cells through the recruitment of CBP to the INSM1 promoter and increase the acetylation of the INSM1 promoter region.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Carrier Proteins/metabolism , DNA-Binding Proteins/genetics , Histones/metabolism , Nerve Tissue Proteins/metabolism , Repressor Proteins/genetics , Transcription Factors/genetics , Acetylation , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Carrier Proteins/genetics , Cell Line , Corticosterone , DNA Primers/genetics , Gene Expression Regulation , Histones/chemistry , Humans , Mice , NIH 3T3 Cells , Nerve Tissue Proteins/genetics , Promoter Regions, Genetic , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection
20.
Pancreas ; 34(2): 248-53, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17312465

ABSTRACT

OBJECTIVES: Islet-associated transcription factors play a critical role in regulating pancreatic endocrine cell differentiation and islet hormone gene expression. Both alpha- and beta-cells differentiate from a common endocrine precursor cell. Therefore, it is important to reveal the differential gene expression profiles between alpha- and beta-cells that can direct their terminal cell fates. alpha-TC-1 and beta-TC-1 are 2 terminally differentiated islet tumor cell lines derived from islets transformed by promoter-specific driven SV40 T antigen overexpression. In this study, we demonstrated that Pdx-1 is a potent transcriptional regulator of endogenous insulin gene expression in alpha-TC-1 cells. METHODS: Reverse transcriptase-polymerase chain reaction and chromatin immunoprecipitation assays were used to analyze gene expression patterns and chromatin modifications in the insulin promoter. RESULTS: Differential transcription factor expression profiles of alpha-TC-1 and beta-TC-1 cells revealed that INSM-1 and Pdx-1 transcription factors were expressed exclusively in beta-TC-1 cells. Overexpression of Ad-Pdx-1 in alpha-TC-1 cells induced insulin gene expression. Chromatin immunoprecipitation assays in Ad-Pdx-1 alpha-TC-1 cells demonstrated Pdx-1 occupancy and the hyperacetylation of histone H4 in the insulin promoter region. CONCLUSIONS: Collectively, these experiments revealed that Pdx-1 is a potent transcriptional regulator that is capable of modulating histone H4 acetylation and activates the insulin gene in a terminally differentiated glucagonoma cell line.


Subject(s)
Glucagon-Secreting Cells/physiology , Histones/metabolism , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/physiology , Insulin/genetics , Trans-Activators/metabolism , Acetylation , Animals , Cell Differentiation , Cell Line, Tumor , Gene Expression/physiology , Glucagon-Secreting Cells/cytology , Homeodomain Proteins/genetics , Insulin-Secreting Cells/cytology , Mice , Mice, Transgenic , Pancreatic Neoplasms , Polymerase Chain Reaction , Promoter Regions, Genetic/physiology , Trans-Activators/genetics , Transcription Factors/genetics , Transcription, Genetic/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...