Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Head Face Med ; 18(1): 9, 2022 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-35236383

RESUMO

BACKGROUND: Stage-specific embryonic antigen-4 (SSEA-4) is a marker for the identification of multipotent embryonic cells. It is also positive in neuroepithelial cells, precursor neural cells (NPC), and human dental pulp cells. The aim of this study was to evaluate the potential morphodifferentiation and histodifferentiation to NPC of SSEA-4 positive stem cells from human exfoliated deciduous teeth (SHED). METHODS: A SHED population in culture, positive to SSEA-4, was obtained by magnetic cell separation. The cells were characterized by immunohistochemistry and flow cytometry. Subsequently, a neurosphere assay was performed in a medium supplemented with basic fibroblast growth factor (bFGF) and epidermal growth factor (EGF); afterward, cells were neurodifferenciated with a neurobasal medium. Finally, indirect immunohistochemistry was performed to identify neuronal markers. RESULTS: The morphological and histological changes in the SSEA-4 positive SHEDs were observed after induction with epidermal and fibroblast growth factors in neurobasal culture medium. At the end of induction, the markers Nestin, TuJ-1, and GFAP were identified. CONCLUSIONS: The findings show that SSEA-4 positive SHEDs have a behavior similar to neuronal precursor cells. Our findings indicate that the dental pulp of deciduous teeth is a promising source for regeneration therapies associated with neurodegenerative diseases or peripheral nerve alterations.


Assuntos
Polpa Dentária , Células-Tronco Neurais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Humanos , Antígenos Embrionários Estágio-Específicos , Dente Decíduo
2.
J BUON ; 25(1): 543-548, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32277681

RESUMO

PURPOSE: To compare the antitumor effect of adenoviruses that express mutant variants of the protein E7 from HPV-16 fused to calreticulin. METHODS: Recombinant adenoviruses were generated to express calreticulin fused to mutant versions of E7 (CRT/E7m and CRT/E7dm). Western blot and immunofluorescence assays were made to demonstrate protein expression. Antitumor assays were performed in C57BL6 mice injected with TC-1 cell line. RESULTS: When HEK293 cells were infected with these adenoviruses, we detected that all the recombinant proteins were expressed at endoplasmic reticulum, as expected. Next, the antitumor effect was tested on a murine tumor model established by inoculation of TC-1 cell line. We detected that both Ad CRT/E7m and Ad CRT/E7dm were capable of reducing the antitumor volume when compared to Ad LacZ, which was used as negative control. No significant difference was observed when compared to Ad CRT/E7, a positive control. CONCLUSIONS: Here we demonstrated that the mutant versions of E7 HPV-16 fused to calreticulin generate similar antitumor effect than the wild type version.


Assuntos
Adenoviridae/patogenicidade , Calreticulina/uso terapêutico , Papillomavirus Humano 16/patogenicidade , Proteínas E7 de Papillomavirus/metabolismo , Animais , Calreticulina/farmacologia , Feminino , Humanos , Camundongos
3.
Cell Stress Chaperones ; 24(1): 149-158, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30604352

RESUMO

Very promising results have been observed with a deoxyribonucleic acid (DNA) vaccine based on human papillomavirus type-16 (HPV-16) antigen retention and delivery system in the endoplasmic reticulum (ER). However, the mechanism by which these antigens are processed once they reach this organelle is still unknown. Therefore, we evaluated whether this system awakens a stress response in the ER. Different DNA constructs based on E6 and E7 mutant antigens fused to an ER signal peptide (SP), a signal for retention in the ER (KDEL), or both signals (SPK), were transfected into HEK-293 cells. Overexpression of E6 and E7 antigens targeted to the ER (SP, and SPK constructs) induced ER stress, which was indicated by an increase of the ER-stress markers GRP78/BiP and CHOP. Additionally, the ER stress response was mediated by the ATF4 transcription factor, which was translocated into the nucleus. Besides, the overexpressed antigens were degraded by the proteasome. Through a cycloheximide-chase assay, we demonstrated that when both protein synthesis and proteasome were inhibited, the overexpressed antigens were degraded. Interestingly, when proteasome was blocked autophagy was increased and the ER stress response decreased. Taken together, these results indicate that the antigens are initially degraded by the ERAD pathway, and autophagy degradation pathway can be induced to compensate the proteasome inhibition. Therefore, we provided a new insight into the mechanism by which E6 and E7 mutant antigens are processed once they reach the ER, which will help to improve the development of more effective vaccines against cancer.


Assuntos
Antígenos Virais/metabolismo , Estresse do Retículo Endoplasmático , Retículo Endoplasmático/metabolismo , Papillomavirus Humano 16/metabolismo , Autofagia , Biomarcadores/metabolismo , Chaperona BiP do Retículo Endoplasmático , Células HEK293 , Humanos , Chaperonas Moleculares/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo
4.
Head Face Med ; 11: 29, 2015 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-26334535

RESUMO

INTRODUCTION: Epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF) play an important role in extracellular matrix mineralization, a complex process required for proper bone regeneration, one of the biggest challenges in dentistry. The purpose of this study was to evaluate the osteogenic potential of EGF and bFGF on dental pulp stem cells (DPSCs). MATERIAL AND METHODS: Human DPSCs were isolated using CD105 magnetic microbeads and characterized by flow cytometry. To induce osteoblast differentiation, the cells were cultured in osteogenic medium supplemented with EGF or bFGF at a low concentration. Cell morphology and expression of CD146 and CD10 surface markers were analyzed using fluorescence microscopy. To measure mineralization, an alizarin red S assay was performed and typical markers of osteoblastic phenotype were evaluated by RT-PCR. RESULTS: EGF treatment induced morphological changes and suppression of CD146 and CD10 markers. Additionally, the cells were capable of producing calcium deposits and increasing the mRNA expression to alkaline phosphatase (ALP) and osteocalcin (OCN) in relation to control groups (p < 0.001). However, bFGF treatment showed an inhibitory effect. CONCLUSION: These data suggests that DPSCs in combination with EGF could be an effective stem cell-based therapy for bone tissue engineering applications in periodontics and oral implantology.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Polpa Dentária/citologia , Família de Proteínas EGF/farmacologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Osteogênese/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Antígeno CD146/metabolismo , Células Cultivadas , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Neprilisina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/metabolismo
5.
Biotechnol Lett ; 37(4): 779-85, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25515795

RESUMO

Fusokines are proteins formed by the fusion of two cytokines. They have greater bioavailability and therapeutic potential than individual cytokines or a combination of different cytokines. Interferon-gamma-inducible protein 10 (CXCL10) and lymphotactin (XCL1) are members of the chemotactic family of cytokines, which induce tumor regression by eliciting immune-system cell chemotaxis. We engineered a replication-deficient adenoviral system expressing CXCL10/XCL1 fusokine (Ad FIL) and assessed its chemotactic response in vitro and in vivo. The CXCL10/XCL1 fusokine elicited a greater chemotactic effect in IL-2 stimulated lymphocytes than individual or combined cytokines in vitro. CXCL10/XCL1 fusokine biological activity was demonstrated in vivo by intratumoral chemoattraction of CXCR3+ cells. Thus, this novel CXCL10/XCL1 fusokine may represent a potential tool for gene therapy treatment of cancer and other illnesses that require triggering immune-system cell recruitment.


Assuntos
Quimiocina CXCL10/metabolismo , Quimiocinas C/metabolismo , Quimiotaxia , Vetores Genéticos , Linfócitos/fisiologia , Mastadenovirus/genética , Animais , Linhagem Celular , Quimiocina CXCL10/genética , Quimiocinas C/genética , Humanos , Linfócitos/efeitos dos fármacos , Camundongos Endogâmicos C57BL
6.
Mol Cancer Ther ; 13(11): 2583-94, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25193509

RESUMO

Pancreatic stellate cells (PSC) have been recognized as the principal cells responsible for the production of fibrosis in pancreatic ductal adenocarcinoma (PDAC). Recently, PSCs have been noted to share characteristics with cells of monocyte-macrophage lineage (MML cells). Thus, we tested whether PSCs could be targeted with the nitrogen-containing bisphosphonates (NBP; pamidronate or zoledronic acid), which are potent MML cell inhibitors. In addition, we tested NBPs treatment combination with nanoparticle albumin-bound paclitaxel (nab-paclitaxel) to enhance antitumor activity. In vitro, we observed that PSCs possess α-naphthyl butyrate esterase (ANBE) enzyme activity, a specific marker of MML cells. Moreover, NBPs inhibited PSCs proliferation, activation, release of macrophage chemoattractant protein-1 (MCP-1), and type I collagen expression. NBPs also induced PSCs apoptosis and cell-cycle arrest in the G1 phase. In vivo, NBPs inactivated PSCs; reduced fibrosis; inhibited tumor volume, tumor weight, peritoneal dissemination, angiogenesis, and cell proliferation; and increased apoptosis in an orthotopic murine model of PDAC. These in vivo antitumor effects were enhanced when NBPs were combined with nab-paclitaxel but not gemcitabine. Our study suggests that targeting PSCs and tumor cells with NBPs in combination with nab-paclitaxel may be a novel therapeutic approach to PDAC.


Assuntos
Carcinoma Ductal Pancreático/tratamento farmacológico , Difosfonatos/farmacologia , Nanopartículas/administração & dosagem , Paclitaxel/administração & dosagem , Neoplasias Pancreáticas/tratamento farmacológico , Células Estreladas do Pâncreas/efeitos dos fármacos , Adulto , Albuminas/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Difosfonatos/administração & dosagem , Sinergismo Farmacológico , Feminino , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Nus , Pamidronato , Neoplasias Pancreáticas/patologia , Células Estreladas do Pâncreas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Oncol Rep ; 30(3): 1506-10, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23783392

RESUMO

Elevated cyclooxygenase-2 (COX-2) expression in breast tumors is associated with a lower survival rate in patients with estrogen receptor α (ERα)-positive tumors. We hypothesized that COX-2 reduces the survival rate of breast cancer patients with ERα-positive tumors since COX-2 increases the invasiveness of ERα-positive breast tumors and decreases tumor sensitivity to tamoxifen. Previously, we demonstrated that COX-2 stimulates the activity of protein kinase C (PKC) to increase the invasiveness of ERα-positive MCF-7 breast cancer cells and to decrease the sensitivity of MCF-7 cells to tamoxifen. High levels of COX-2 are associated with the activation of the mitogen-activated protein kinase (MAPK) family and the Akt kinase. However, it is not known whether these kinases mediate COX-2-induced invasive activity and tamoxifen resistance. In the present study, we report that COX-2 utilizes PKC to enhance the phosphorylation of Jun N-terminal kinases (JNKs), but not that of other MAPK family members or Akt. Inhibition aimed at JNKs reduced COX-2-induced invasion but not COX-2-induced tamoxifen resistance. We conclude that JNKs are essential for induced cell invasion by COX-2, but not tamoxifen resistance, in ERα-positive breast cancer cells.


Assuntos
Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Ciclo-Oxigenase 2/metabolismo , Resistencia a Medicamentos Antineoplásicos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Tamoxifeno/farmacologia , Apoptose , Western Blotting , Neoplasias da Mama/metabolismo , Movimento Celular , Proliferação de Células , Colágeno/metabolismo , Combinação de Medicamentos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Laminina/metabolismo , Células MCF-7 , Invasividade Neoplásica , Proteínas Nucleares/metabolismo , Fosforilação/efeitos dos fármacos , Proteína Quinase C/metabolismo , Proteoglicanas/metabolismo , Transdução de Sinais/efeitos dos fármacos
8.
Oncol Rep ; 27(3): 861-6, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22200873

RESUMO

Obesity is a significant risk factor for post-menopausal women to develop and die from breast cancer. Leptin, an adipokine is produced in high levels in obese individuals, and its receptor is overexpressed in breast tumors and lymph node metastases. Previously, we demonstrated that leptin stimulates breast cancer cell invasion, which is correlated with breast cancer metastasis. Programmed cell death 4 (PDCD4) has been shown to block cancer cell invasion. However, whether PDCD4 blocks leptin-induced breast cancer cell invasion is not known. Here, we report the novel findings that leptin failed to induce invasion in MCF-7 breast cancer cells overexpressing PDCD4 (MCF-7/PDCD4). Tissue inhibitor of metalloproteinase-2 (TIMP-2) was essential to the anti-invasive effect of PDCD4, as leptin stimulated the invasion of MCF-7/PDCD4 cells pretreated with TIMP-2 siRNA. Furthermore, TIMP-2 knockdown allowed leptin to augment phosphorylation of extracellular signal-regulated kinases 1,2 and signal transducer and activator of transcription 3, but not that of Jun N-terminal kinases. These data indicate that PDCD4 utilizes TIMP-2 to exert its anti-invasive effect by suppressing leptin-induced activation of extracellular signal-regulated kinases 1,2 and signal transducer and activator of transcription 3. Novel therapeutic strategies aiming at enhancing PDCD4 expression in breast tumors may be able to stop obesity-related breast tumor progression and prolong the life of patients.


Assuntos
Proteínas Reguladoras de Apoptose/biossíntese , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Leptina/antagonistas & inibidores , Proteínas de Ligação a RNA/biossíntese , Proteínas Reguladoras de Apoptose/genética , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Feminino , Técnicas de Silenciamento de Genes/métodos , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Leptina/metabolismo , Leptina/farmacologia , Sistema de Sinalização das MAP Quinases , Invasividade Neoplásica , Fosforilação/genética , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/genética , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Inibidor Tecidual de Metaloproteinase-2/genética , Inibidor Tecidual de Metaloproteinase-2/metabolismo , Transfecção
9.
Biotechnol Lett ; 31(2): 215-20, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18923910

RESUMO

Lymphotactin-XCL1 is a chemokine produced mainly by activated CD8+ T-cells and directs migration of CD4+ and CD8+ lymphocytes and natural killer (NK) cells. We expressed human lymphotactin (LTN) by the lactic-acid bacterium Lactococcus lactis. Biological activity of LTN was confirmed by chemo-attraction of human T-cells by chemotaxis demonstrating, for the first time, how this chemokine secreted by a food-grade prokaryote retains biological activity and chemoattracts T lymphocytes. This strain thus represents a feasible well-tolerated vector to deliver active LTN at a mucosal level.


Assuntos
Quimiocinas C/biossíntese , Quimiocinas C/farmacologia , Quimiotaxia/fisiologia , Lactococcus lactis/metabolismo , Linfócitos/efeitos dos fármacos , Linfócitos/fisiologia , Engenharia de Proteínas/métodos , Células Cultivadas , Quimiocinas C/genética , Quimiotaxia/efeitos dos fármacos , Humanos , Lactococcus lactis/genética , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/metabolismo
10.
Microb Cell Fact ; 7: 22, 2008 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-18662403

RESUMO

BACKGROUND: Chemokines are a large group of chemotactic cytokines that regulate and direct migration of leukocytes, activate inflammatory responses, and are involved in many other functions including regulation of tumor development. Interferon-gamma inducible-protein-10 (IP-10) is a member of the C-X-C subfamily of the chemokine family of cytokines. IP-10 specifically chemoattracts activated T lymphocytes, monocytes, and NK cells. IP-10 has been described also as a modulator of other antitumor cytokines. These properties make IP-10 a novel therapeutic molecule for the treatment of chronic and infectious diseases. Currently there are no suitable live biological systems to produce and secrete IP-10. Lactococcus lactis has been well-characterized over the years as a safe microorganism to produce heterologous proteins and to be used as a safe, live vaccine to deliver antigens and cytokines of interest. Here we report a recombinant strain of L. lactis genetically modified to produce and secrete biologically active IP-10. RESULTS: The IP-10 coding region was isolated from human cDNA and cloned into an L. lactis expression plasmid under the regulation of the pNis promoter. By fusion to the usp45 secretion signal, IP-10 was addressed out of the cell. Western blot analysis demonstrated that recombinant strains of L. lactis secrete IP-10 into the culture medium. Neither degradation nor incomplete forms of IP-10 were detected in the cell or supernatant fractions of L. lactis. In addition, we demonstrated that the NICE (nisin-controlled gene expression) system was able to express IP-10 "de novo" even two hours after nisin removal. This human IP-10 protein secreted by L. lactis was biological active as demonstrated by Chemotaxis assay over human CD3+T lymphocytes. CONCLUSION: Expression and secretion of mature IP-10 was efficiently achieved by L. lactis forming an effective system to produce IP-10. This recombinant IP-10 is biologically active as demonstrated by its ability to chemoattract human CD3+ T lymphocytes. This strain of recombinant L. lactis represents a potentially useful tool to be used as a live vaccine in vivo.

11.
Int J Oncol ; 31(5): 1243-50, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17912453

RESUMO

We previously demonstrated that liposome-incorporated antisense oligodeoxynucleotide specific for the grb2 mRNA (L-Grb2) inhibited Grb2 protein expression and the proliferation of bcr-abl-positive leukemia cell lines. To determine whether L-Grb2 has the potential of being a therapeutic modality against bcr-abl-positive leukemia, we studied the tissue distribution of L-Grb2 in normal mice before studying its effects in mice bearing bcr-abl-positive leukemia xenografts. L-Grb2 was widely distributed in the body. The highest tissue concentrations of L-Grb2 were found in the spleen and liver, which are the organs where the tumor mass of bcr-abl-positive leukemia is mainly found. At 4 h post-injection, the amount of L-Grb2 detected per g of tissue was 64 microg in spleen and 50 microg in liver. Intravenous injection of bcr-abl-positive 32D mouse leukemia cells into radiated NOD/scid mice caused a lethal leukemia syndrome; we determined whether L-Grb2 could prolong the survival of mice bearing such xenografts. One day after leukemia cell inoculation, mice received twice weekly intravenous injections of L-Grb2. At an injection dose of 15 mg of L-Grb2 per kg of mouse body weight, 80% of mice treated with L-Grb2 survived to 48 days (end of study) whereas 0% of mice treated with the same dose of liposomal control oligonucleotide survived; the mean survival duration of these groups was 44 and 20 days, respectively. Our data indicate that L-Grb2 prolonged the survival of mice bearing bcr-abl-positive leukemia xenografts. L-Grb2 may be used as a novel cancer therapeutic modality.


Assuntos
Proteínas de Fusão bcr-abl/análise , Proteína Adaptadora GRB2/antagonistas & inibidores , Leucemia Experimental/terapia , Oligodesoxirribonucleotídeos Antissenso/administração & dosagem , Animais , Proteína Adaptadora GRB2/genética , Leucemia Experimental/mortalidade , Contagem de Leucócitos , Lipossomos , Camundongos , Camundongos Endogâmicos ICR , Transplante de Neoplasias , Oligodesoxirribonucleotídeos Antissenso/farmacocinética , Ratos , Ratos Endogâmicos Lew , Distribuição Tecidual , Transplante Heterólogo
12.
Lab Invest ; 85(11): 1357-67, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16127422

RESUMO

Approximately 30-40% of estrogen receptor alpha (ERalpha)-positive breast tumors express high levels of the cyclooxygenase-2 (COX-2) protein, and these high levels have been associated with a poorer prognosis in breast cancer patients. We speculate that high levels of COX-2 induce drug resistance in ERalpha-positive breast tumors, thus reducing the survival rate of patients with such tumors. Human breast cancer cell lines that express high levels of COX-2 are generally ERalpha negative. To determine whether COX-2 induces drug resistance, plasmids encoding the COX-2 gene were stably transfected into ERalpha-positive MCF-7 human breast cancer cells (MCF-7/COX-2). MCF-7/COX-2 cells were resistant to the selective estrogen receptor modulator tamoxifen but not to its analog, raloxifene. MCF-7/COX-2 cells were also resistant to the retinoid N-(4-hydroxyphenyl)retinamide (4-HPR) but not to its analog, all-trans retinoic acid. In contrast, the sensitivities of MCF-7/COX-2 cells to doxorubicin and paclitaxel were similar to those of the parental MCF-7 cells. We then determined which COX-2 product, prostaglandin E2 (PGE2) or prostaglandin F2alpha is involved in the COX-2-mediated drug resistance. PGE2, but not PGF2alpha, blocked the antiproliferative effects of tamoxifen and 4-HPR. Agonists that activate PGE2 receptors and their downstream kinase effectors, protein kinases A and C, also blocked the growth inhibitory effects of these drugs. Increased levels of Bcl-2 and Bcl-XL proteins have been reported in mammary tumors of COX-2 transgenic mice and in human colon cancer cell lines that have high levels of COX-2. However, we did not observe any changes in Bcl-2, Bcl-XL, or Bax expression induced by COX-2 or PGE2. Here we report the novel findings that COX-2 uses PGE2 to stimulate the activities of protein kinases A and C to induce selectively tamoxifen and 4-HPR resistance in ERalpha-positive breast cancer cells.


Assuntos
Antineoplásicos/metabolismo , Ciclo-Oxigenase 2/metabolismo , Fenretinida/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Tamoxifeno/antagonistas & inibidores , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Células Clonais , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Concentração Inibidora 50
13.
Leuk Lymphoma ; 44(11): 1979-85, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14738153

RESUMO

A P-ethoxy oligonucleotide (oligo), 20 bases long and specific for the translation initiation site of human Bcl-2 mRNA, was incorporated into liposomes to increase its intracellular delivery. This oligo selectively inhibited Bcl-2 protein expression and induced growth inhibition in t(14;18)-positive transformed follicular lymphoma (FL) cell lines. We studied the inhibitory effects of shorter liposomal P-ethoxy oligos (7, 9, 11 or 15 mer) in order to determine the activity of different oligo chain lengths targeted to the same Bcl-2 mRNA. At 12 microM, all the oligos inhibited the growth of a FL cell line. We compared the 7-mer oligo with the 20-mer oligo. The two oligos inhibited Bcl-2 protein expression similarly: 66% and 60% for the 7- and 20-mer, respectively. The uptake and retention of both oligos were also very similar. Our results indicate that the Bcl-2 inhibitory activity is maintained with P-ethoxy antisense oligos ranging from 7 to 20 bases.


Assuntos
Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Linfoma Folicular/patologia , Oligonucleotídeos Antissenso/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA Mensageiro/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Transformação Celular Neoplásica , Regulação para Baixo , Sistemas de Liberação de Medicamentos , Etil-Éteres/farmacologia , Marcação de Genes , Humanos , Lipossomos , Linfoma Folicular/genética , Células Tumorais Cultivadas
14.
Semin Oncol ; 29(3 Suppl 11): 71-6, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12138400

RESUMO

High expression of the bcl-2 proto-oncogene is found in various human hematologic malignancies and solid tumors. Bcl-2 protein exerts its oncogenic role by preventing tumor cells from undergoing apoptosis induced by radiation, chemotherapy, and hormonal therapy. Antisense oligonucleotides directed toward the open reading frame of the bcl-2 gene have been used to inhibit Bcl-2 expression. Inhibition of Bcl-2 expression sensitizes lymphoma and leukemia cells to radiation and chemotherapy. However, it remains to be determined whether Bcl-2 antisense oligonucleotides will have a beneficial effect in solid tumors, such as breast cancer. Laboratory results indicate that Bcl-2 overexpression induces endocrine and chemoresistance in breast cancer cells. However, high levels of Bcl-2 have been associated with favorable prognostic factors, suggesting that Bcl-2 may not be an appropriate target in breast cancer. We will discuss the paradoxical role of Bcl-2 and the potential therapeutic application of Bcl-2 antisense oligonucleotides in breast cancer.


Assuntos
Neoplasias da Mama/terapia , Oligodesoxirribonucleotídeos Antissenso/uso terapêutico , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Apoptose/genética , Neoplasias da Mama/genética , Resistencia a Medicamentos Antineoplásicos/fisiologia , Feminino , Regulação Neoplásica da Expressão Gênica , Genes bcl-2/fisiologia , Humanos , Proto-Oncogene Mas
15.
Tumour Biol ; 23(5): 279-86, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12595744

RESUMO

We previously reported that overexpression of the HER2/NEU oncogene induces all-TRANS retinoic acid (ATRA) resistance in breast cancer cells. N-(4-hydroxyphenyl)-retinamide (4HPR), a synthetic analogue of ATRA, has been shown to repress the expression of HER2/neu and its family member, epidermal growth factor receptor (EGFR). We investigated whether 4HPR, by suppressing HER2/neu or EGFR expression, could sensitize breast cancer cells to ATRA. At 1.3 micro M concentration (a clinically pharmacologically achievable dose), 4HPR increased ATRA sensitivity synergistically in HER2/NEU-overexpressing BT-474, MDA-MB-453, and MCF-7/Her2 breast cancer cells. However, 4HPR did not sensitize EGFR-overexpressing MDA-MB-468, Hs578T, and MCF-7/EGFR breast cancer cells to ATRA. The increased inhibitory effects in HER2/NEU-overexpressing cells were not correlated with increases in expression levels of p21(WAF1/CIP1) or retinoblastoma protein. Combining 4HPR with ATRA may lead to a novel, selective therapeutic or chemopreventive strategy against HER2/NEU-overexpressing breast tumors.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Fenretinida/farmacologia , Receptor ErbB-2/análise , Tretinoína/farmacologia , Neoplasias da Mama/química , Neoplasias da Mama/patologia , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/análise , Sinergismo Farmacológico , Receptores ErbB/análise , Feminino , Humanos , Proteína do Retinoblastoma/análise , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA