Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Biology (Basel) ; 12(8)2023 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-37627018

RESUMO

Human neuroblastoma (NB) is the most common childhood extracranial tumor arising from the sympathetic nervous system. It is also a clinically heterogeneous disease that ranges from spontaneous regression to high-risk stage 4 disease. The cause of this disease remains elusive. However, the amplification of NMYC oncogene occurred in roughly 30% of NB patients, which strongly correlated with the advanced stage of disease subtype and the worse prognosis status. We discovered that N-Myc oncoprotein binds and activates INSM1, a zinc-finger transcription factor of neuroendocrine tumors. We also found that INSM1 modulates N-Myc stability mediated through PI3K/AKT/GSK3ß signaling pathway. Therefore, INSM1 emerges as a critical co-player with N-Myc in facilitating NB tumor cell growth and sustaining the advanced stage of malignancy. Using an INSM1-promoter driven luciferase screening-platform, we have recently identified fifteen small molecules that negatively regulate INSM1 expression. Interestingly, the identified small molecules can be divided into four large groups of compounds such as cell signaling inhibitor, DNA/RNA inhibitor, HDAC inhibitor, and cardiac glycoside. These findings support the presence of a unique mechanism associated with INSM1 and N-Myc interplay, which is critical in regulating NB tumor cell growth. We discuss the feasibility of identifying novel or repurposing small molecules targeting INSM1 as a potential treatment option for high-risk NB.

2.
Cell Signal ; 109: 110753, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37301315

RESUMO

High-risk neuroblastoma (NB) is a heterogeneous and malignant childhood cancer that is frequently characterized by MYCN proto-oncogene amplification or elevated N-Myc protein (N-Myc) expression. An N-Myc downstream target gene, insulinoma associated-1 (INSM1) has emerged as a biomarker that plays a critical role in facilitating NB tumor cell growth and transformation. N-Myc activates endogenous INSM1 gene expression through binding to the E2-box of the INSM1 proximal promoter in NB. We identified a plant alkaloid, homoharringtonine (HHT), from a chemical library screening showing potent inhibition of INSM1 promoter activity. This positive-hit plant alkaloid exemplifies an effective screening approach for repurposed compound targeting INSM1 expression in NB cancer therapy. The elevated N-Myc and INSM1 expression in NB constitutes a positive-loop through INSM1 activation that promotes N-Myc stability. In the present study, the biological effects and anti-tumor properties of HHT against NB were examined. HHT either down regulates and/or interferes with the binding of N-Myc to the E2-box of the INSM1 promoter and the inhibition of PI3K/AKT-mediated N-Myc stability could lead to the NB cell apoptosis. HHT inhibition of NB cell proliferation is consistent with the INSM1 expression as higher level of INSM1 exhibits a more sensitive IC50 value. The combination treatment of HHT and A674563 provides a better option of increasing potency and reducing cellular cytotoxicity than HHT or A674563 treatment alone. Taken together, the suppression of the INSM1-associated signaling pathway axis promotes the inhibition of NB tumor cell growth. This study developed a feasible approach for repurposing an effective anti-NB drug.


Assuntos
Insulinoma , Neuroblastoma , Neoplasias Pancreáticas , Humanos , Criança , Mepesuccinato de Omacetaxina , Fosfatidilinositol 3-Quinases/metabolismo , Reposicionamento de Medicamentos , Linhagem Celular Tumoral , Neuroblastoma/genética , Neoplasias Pancreáticas/genética , Regulação Neoplásica da Expressão Gênica , Proteínas Repressoras/metabolismo
3.
Biology (Basel) ; 11(10)2022 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-36290282

RESUMO

An aggressive form of neuroblastoma (NB), a malignant childhood cancer derived from granule neuron precursors and sympathoadrenal lineage, frequently comprises MYCN amplification/elevated N-Myc expression, which contributes to the development of neural crest-derived embryonal malignancy. N-Myc is an oncogenic driver in NB. Persistent N-Myc expression during the maturation of SA precursor cells can cause blockage of the apoptosis and induce abnormal proliferation, resulting in NB development. An insulinoma-associated-1 (INSM1) zinc-finger transcription factor has emerged as an NB biomarker that plays a critical role in facilitating tumor cell growth and transformation. INSM1 plays an essential role in sympathoadrenal cell differentiation. N-Myc activates endogenous INSM1 through an E2-box of the INSM1 proximal promoter, whereas INSM1 enhances N-Myc stability via RAC-α-serine/threonine protein kinase (AKT) phosphorylation in NB. The ectopic expression of INSM1 stimulates NB tumor growth in contrast to the knockdown of INSM1 that inhibits NB cell proliferation. The clinical pathological result and bioinformatics analysis show that INSM1 is a strong diagnostic and a prognostic biomarker for the evaluation of NB progression. The INSM1/N-Myc expression shows high clinical relevance in NB. Therefore, targeting the INSM1/N-Myc-associated signaling axis should be a feasible approach to identifying new drugs for the suppression of NB tumor growth.

4.
Cell Signal ; 76: 109785, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32966884

RESUMO

Aggressive form of neuroblastoma (NB) is a malignant childhood cancer derived from granule neuron precursors and sympatho-adrenal lineage with N-MYC amplification. An insulinoma associated-1 (INSM1) transcription factor has emerged as a NB biomarker that plays critical role in facilitating tumor cell growth and transformation. N-myc activates INSM1 in NB was discovered. In order to elucidate the signaling pathways associated with INSM1 expression and NB tumor cell growth, we developed an INSM1 promoter-driven luciferase assay for new drug discovery. Promoter-driven luciferase assay demonstrated high Z' factor (>0.7). Performance measures using signal-to-noise ratio, signal window, and Z' factor confirmed this assay is a robust screening method. A panel of FDA-approved oncology drugs set (147 compounds) was subjected to the INSM1 promoter-driven luciferase assay. The positive-hit compounds were validated for effects on cell viability and INSM1 expression. Screening a FDA-approved oncology drugs set revealed multiple inhibitors against various signaling pathways via suppression of INSM1 promoter activity. The positive-hit compounds consist of 9 signaling pathway inhibitors negatively regulates INSM1 expression and cell viability in NB. These inhibitors target DNA/RNA/protein synthesis, tubulin assembly, and histone deacetylase signaling pathways. The outcome of this assay indicates that the newly identified pathways could be critical for NB growth and transformation. This technology and the positive-hits of multiple pathways represent a promising option of identifying re-purposed FDA-approved drugs valuable for NB treatment in the context of a NB-specific marker, INSM1.


Assuntos
Antineoplásicos/farmacologia , Reposicionamento de Medicamentos/métodos , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Neuroblastoma/tratamento farmacológico , Proteínas Repressoras/antagonistas & inibidores , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos
5.
Mol Cancer Res ; 17(8): 1597-1604, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31113827

RESUMO

Insulinoma-associated-1 (IA-1 or INSM1) encodes a zinc-finger transcription factor, which was isolated from a human insulinoma subtraction library, with specific expression patterns, predominantly in developing neuroendocrine tissues and tumors. INSM1 is key in early pancreatic endocrine, sympatho-adrenal lineage, and pan-neurogenic precursor development. Insm1 gene ablation results in impairment of pancreatic ß cells, catecholamine biosynthesis, and basal progenitor development during mammalian neocortex maturation. Recently, INSM1 has emerged as a superior, sensitive, and specific biomarker for neuroendocrine tumors. INSM1 regulates downstream target genes and exhibits extranuclear activities associated with multiple signaling pathways, including Sonic Hedgehog, PI3K/AKT, MEK/ERK1/2, ADK, p53, Wnt, histone acetylation, LSD1, cyclin D1, Ascl1, and N-myc. Novel strategies targeting INSM1-associated signaling pathways facilitate the suppression of neuroendocrine tumor growth. In addition, INSM1 promoter-driven reporter assay and/or suicide gene therapy are promising effective therapeutic approaches for targeted specific neuroendocrine tumor therapy. In this review, the current knowledge of the biological role of INSM1 as a neuroendocrine tumor biomarker is summarized, and novel strategies targeting multiple signaling pathways in the context of INSM1 expression in neuroendocrine tumors are further explored. IMPLICATIONS: Neuroendocrine transcription factor (INSM1) may serve as a neuroendocrine biomarker for the development of novel cancer therapeutics against neuroendocrine tumors.


Assuntos
Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/metabolismo , Terapia de Alvo Molecular , Tumores Neuroendócrinos/diagnóstico , Proteínas Repressoras/metabolismo , Biomarcadores Tumorais/genética , Humanos , Tumores Neuroendócrinos/tratamento farmacológico , Tumores Neuroendócrinos/metabolismo , Proteínas Repressoras/genética , Transdução de Sinais
6.
J Biol Chem ; 294(14): 5456-5465, 2019 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-30755485

RESUMO

Insulinoma-associated-1 (INSM1) is a key protein functioning as a transcriptional repressor in neuroendocrine differentiation and is activated by N-Myc in human neuroblastoma (NB). INSM1 modulates the phosphoinositide 3-kinase (PI3K)-AKT Ser/Thr kinase (AKT)-glycogen synthase kinase 3ß (GSK3ß) signaling pathway through a positive-feedback loop, resulting in N-Myc stabilization. Accordingly, INSM1 has emerged as a critical player closely associated with N-Myc in facilitating NB cell growth. Here, an INSM1 promoter-driven luciferase-based screen revealed that the compound 5'-iodotubercidin suppresses adenosine kinase (ADK), an energy pathway enzyme, and also INSM1 expression and NB tumor growth. Next, we sought to dissect how the ADK pathway contributes to NB tumor cell growth in the context of INSM1 expression. We also found that 5'-iodotubercidin inhibits INSM1 expression and induces an intra- and extracellular adenosine imbalance. The adenosine imbalance, which triggers adenosine receptor-3 signaling that decreases cAMP levels and AKT phosphorylation and enhances GSK3ß activity. We further observed that GSK3ß then phosphorylates ß-catenin and promotes the cytoplasmic proteasomal degradation pathway. 5'-Iodotubercidin treatment and INSM1 inhibition suppressed extracellular signal-regulated kinase 1/2 (ERK1/2) activity and the AKT signaling pathways required for NB cell proliferation. The 5'-iodotubercidin treatment also suppressed ß-catenin, lymphoid enhancer-binding factor 1 (LEF-1), cyclin D1, N-Myc, and INSM1 levels, ultimately leading to apoptosis via caspase-3 and p53 activation. The identification of the signaling pathways that control the proliferation of aggressive NB reported here suggests new options for combination treatments of NB patients.


Assuntos
Proliferação de Células/efeitos dos fármacos , AMP Cíclico/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas de Neoplasias/metabolismo , Neuroblastoma/tratamento farmacológico , Proteínas Repressoras/biossíntese , Tubercidina/análogos & derivados , Apoptose/efeitos dos fármacos , Humanos , Células K562 , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas de Neoplasias/genética , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Proto-Oncogene Mas , Proteínas Repressoras/genética , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Tubercidina/farmacologia
7.
J Transl Med ; 16(1): 297, 2018 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-30359270

RESUMO

BACKGROUND: Neurogenin3 (Ngn3) and neurogenic differentiation 1 (NeuroD1), two crucial transcriptional factors involved in human diabetes (OMIM: 601724) and islet development, have been previously found to directly target to the E-boxes of the insulinoma-associated 2 (Insm2) gene promoter, thereby activating the expression of Insm2 in insulin-secretion cells. However, little is known about the function of Insm2 in pancreatic islets and glucose metabolisms. METHODS: Homozygous Insm2-/- mice were generated by using the CRISPR-Cas9 method. Glucose-stimulated insulin secretion and islet morphology were analyzed by ELISA and immunostainings. Expression levels of Insm2-associated molecules were measured using quantitative RT-PCR and Western blots. RESULTS: Fasting blood glucose levels of Insm2-/- mice were higher than wild-type counterparts. Insm2-/- mice also showed reduction in glucose tolerance and insulin/C-peptide levels when compared to the wild-type mice. RT-PCR and Western blot analysis revealed that expression of Insm1 was significantly increased in Insm2-/- mice, suggesting a compensatory response of the homolog gene Insm1. Similarly, transcriptional levels of Ngn3 and NeuroD1 were also increased in Insm2-/- mice. Moreover, Insm2-/- female mice showed a significantly decreased reproductive capacity. CONCLUSIONS: Our findings suggest that Insm2 is important in glucose-stimulated insulin secretion and is involved in the development pathway of neuroendocrine tissues which are regulated by the transcription factors Ngn3, NeuroD1 and Insm1.


Assuntos
Deleção de Genes , Intolerância à Glucose/genética , Secreção de Insulina , Fatores de Transcrição/genética , Animais , Sequência de Bases , Feminino , Genótipo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos Knockout , Modelos Biológicos , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Tirosina Fosfatases Classe 8 Semelhantes a Receptores/genética , Proteínas Tirosina Fosfatases Classe 8 Semelhantes a Receptores/metabolismo
8.
Cell Signal ; 46: 83-91, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29501727

RESUMO

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.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas Hedgehog/metabolismo , Proteína Proto-Oncogênica N-Myc/metabolismo , Tumores Neuroendócrinos/metabolismo , Proteínas Repressoras/metabolismo , Carcinoma de Pequenas Células do Pulmão/metabolismo , Homeobox 2 de Ligação a E-box com Dedos de Zinco/metabolismo , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Regiões Promotoras Genéticas/genética , Proteínas Repressoras/genética , Transdução de Sinais
9.
Int J Oncol ; 49(2): 847, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27279381

RESUMO

Following the publication of this article, an interested reader drew to our attention an anomaly associated with the presentation of Figs. 5 and 8; essentially, the ß-actin control bands in Fig. 8 appeared to be a mirror image of the same control bands shown in Fig. 5A. After having re­examined the data, Fig. 5 was correctly presented as shown in the paper, although the ß-actin blot in Fig. 8 had been inadvertently misplaced as a mirror image of the control data from Fig. 5. Two similar experiments were performed at that time, yielding similar results, and the error with the control data did not affect the second of the experiments. Therefore, we present a corrected version of Fig. 8 containing the alternative data. The findings and conclusions of this paper are still supported by our experimental data, and were not affected by this error. We sincerely apologize for this mistake, and thank the reader of our article who drew this matter to our attention. Furthermore, we regret any inconvenience this mistake has caused. [the original article was published in the International Journal of Oncology 42: 1113-1119, 2013; DOI: 10.3892/ijo.2013.1789].

10.
J Biol Chem ; 291(21): 11313-22, 2016 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-27048654

RESUMO

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.


Assuntos
Células Epiteliais/metabolismo , Timo/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Linhagem Celular , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Células Epiteliais/patologia , Humanos , Mutação , Estrutura Terciária de Proteína , Timo/patologia , Fatores de Transcrição/genética , Proteína AIRE
11.
BMC Pulm Med ; 16: 49, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-27072116

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/genética , Expressão Ectópica do Gene/genética , Células Epiteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/metabolismo , Alvéolos Pulmonares/embriologia , Carcinoma de Pequenas Células do Pulmão/metabolismo , Fatores de Transcrição/genética , Animais , Northern Blotting , Western Blotting , Brônquios/citologia , Brônquios/metabolismo , Estudos de Casos e Controles , Linhagem Celular , Ciclina D1/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células Epiteliais/citologia , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Pulmão/embriologia , Pulmão/crescimento & desenvolvimento , Pulmão/metabolismo , Camundongos , Camundongos Transgênicos , Alvéolos Pulmonares/crescimento & desenvolvimento , Alvéolos Pulmonares/metabolismo , Proteínas Repressoras , Mucosa Respiratória/citologia , Mucosa Respiratória/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/metabolismo
12.
Cell Oncol (Dordr) ; 39(3): 279-86, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26902080

RESUMO

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.


Assuntos
Terapia Genética/métodos , Insulinoma , Terapia Viral Oncolítica/métodos , Neoplasias Pancreáticas , Regiões Promotoras Genéticas , Proteínas Repressoras/genética , Adenoviridae , Animais , Antineoplásicos/farmacologia , Western Blotting , Linhagem Celular Tumoral , Ganciclovir/farmacologia , Vetores Genéticos , Humanos , Camundongos , Timidina Quinase/genética , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Int J Oncol ; 48(1): 173-80, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26530405

RESUMO

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.


Assuntos
Terapia Genética , Luciferases/genética , Tumores Neuroendócrinos/genética , Proteínas Repressoras/genética , Adenoviridae/genética , Animais , Carcinoma Neuroendócrino , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Vetores Genéticos , Humanos , Luciferases/biossíntese , Camundongos , Tumores Neuroendócrinos/diagnóstico , Tumores Neuroendócrinos/patologia , Regiões Promotoras Genéticas
14.
Oncotarget ; 6(34): 36700-12, 2015 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-26456864

RESUMO

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.


Assuntos
Proteína Proto-Oncogênica N-Myc/metabolismo , Neuroblastoma/metabolismo , Proteínas Repressoras/metabolismo , Animais , Carcinogênese , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Xenoenxertos , Humanos , Camundongos , Proteína Proto-Oncogênica N-Myc/genética , Neuroblastoma/genética , Neuroblastoma/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Repressoras/genética , Transdução de Sinais , Fatores de Transcrição/metabolismo
15.
Life Sci ; 114(1): 45-50, 2014 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-25128855

RESUMO

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.


Assuntos
Células Endócrinas/citologia , Produtos do Gene tat/genética , Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Fatores de Transcrição/genética , Células Acinares/citologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/genética , Linhagem Celular , Cromatografia de Afinidade , Cricetinae , Vetores Genéticos , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Proteínas do Tecido Nervoso/genética , Regiões Promotoras Genéticas , Ratos , Proteínas Recombinantes/genética , Transativadores/genética , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética
16.
Cell Signal ; 26(4): 740-7, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24407176

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Homeodomínio/metabolismo , Neuropeptídeos/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Cromonas/farmacologia , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Proteínas de Homeodomínio/genética , Insulina/genética , Insulina/metabolismo , Camundongos , Dados de Sequência Molecular , Morfolinas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Regiões Promotoras Genéticas , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Receptor de Insulina/metabolismo , Receptores de Quinase C Ativada , Proteínas Repressoras , Fatores de Transcrição/química , Fatores de Transcrição/genética
17.
Virol J ; 10: 86, 2013 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-23497017

RESUMO

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.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Lentivirus/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Glicoproteínas de Membrana/genética , Transdução Genética/métodos , Proteínas do Envelope Viral/genética , Linhagem Celular Tumoral , Dependovirus/fisiologia , Terapia Genética/instrumentação , Vetores Genéticos/genética , Vetores Genéticos/fisiologia , Humanos , Lentivirus/metabolismo , Neoplasias Pulmonares/virologia , Glicoproteínas de Membrana/metabolismo , Transdução Genética/instrumentação , Proteínas do Envelope Viral/metabolismo
18.
Ochsner J ; 13(1): 91-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23533081

RESUMO

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.

19.
Int J Oncol ; 42(3): 1113-9, 2013 03.
Artigo em Inglês | MEDLINE | ID: mdl-23354006

RESUMO

Obesity is known to be an important risk factor for many types of cancer, such as breast, prostate, liver and endometrial cancer. Recently, epidemiological studies have indicated that obesity correlates with an increased risk of developing ovarian cancer, the most lethal gynecological cancer in developed countries. Leptin is predominantly produced by adipocytes and acts as a growth factor and serum leptin levels positively correlate with the amount of body fat. In this study, we investigated the effects of leptin on the growth of ovarian cancer cells and the underlying mechanism(s) of action. Our results showed that leptin stimulated the growth of the OVCAR-3 ovarian cancer cell line using MTT assay and trypan blue exclusion. Using western blot analysis, we found that leptin enhanced the expression of cyclin D1 and Mcl-1, which are important regulators of cell proliferation and the inhibition of apoptosis. To investigate the signaling pathways that mediate the effects of leptin, cells were treated with leptin plus specific inhibitors of JAK2, PI3K/Akt and MEK/ERK1/2 and analysis of the phosphorylation state of proteins was carried out by western blot assays. We showed that the activation of the MEK/ERK1/2 and PI3K/Akt signaling pathways were involved in the growth-stimulating effect of leptin on ovarian cancer cell growth and the specific inhibitors of PI3K/Akt and MEK/ERK1/2 revealed that these two pathways interacted with each other. Our data demonstrate that leptin upregulates the expression of cyclin D1 and Mcl-1 to stimulate cell growth by activating the PI3K/Akt and MEK/ERK1/2 pathways in ovarian cancer.


Assuntos
Ciclina D1/metabolismo , Leptina/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Neoplasias Ovarianas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Apoptose , Butadienos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Ciclina D1/biossíntese , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Flavonoides/farmacologia , Humanos , Janus Quinase 2/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteína de Sequência 1 de Leucemia de Células Mieloides , Nitrilas/farmacologia , Obesidade/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Tirfostinas/farmacologia
20.
J Cell Physiol ; 227(6): 2470-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21830214

RESUMO

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.


Assuntos
Células Acinares/metabolismo , Transdiferenciação Celular , Ilhotas Pancreáticas/metabolismo , Proteínas Repressoras/metabolismo , Células Acinares/efeitos dos fármacos , Células Acinares/patologia , Adenoviridae/genética , Amilases/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Linhagem Celular Tumoral , Transdiferenciação Celular/efeitos dos fármacos , Cricetinae , Dexametasona/farmacologia , Receptores ErbB/metabolismo , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Vetores Genéticos , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Insulina/genética , Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/patologia , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/metabolismo , RNA Mensageiro/metabolismo , Ratos , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteínas Repressoras/genética , Transativadores/genética , Transativadores/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...