Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Nat Genet ; 55(6): 1022-1033, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37169874

RESUMO

Patients with high-risk neuroblastoma generally present with widely metastatic disease and often relapse despite intensive therapy. As most studies to date focused on diagnosis-relapse pairs, our understanding of the genetic and clonal dynamics of metastatic spread and disease progression remain limited. Here, using genomic profiling of 470 sequential and spatially separated samples from 283 patients, we characterize subtype-specific genetic evolutionary trajectories from diagnosis through progression and end-stage metastatic disease. Clonal tracing timed disease initiation to embryogenesis. Continuous acquisition of structural variants at disease-defining loci (MYCN, TERT, MDM2-CDK4) followed by convergent evolution of mutations targeting shared pathways emerged as the predominant feature of progression. At diagnosis metastatic clones were already established at distant sites where they could stay dormant, only to cause relapses years later and spread via metastasis-to-metastasis and polyclonal seeding after therapy.


Assuntos
Recidiva Local de Neoplasia , Neuroblastoma , Humanos , Recidiva Local de Neoplasia/genética , Neuroblastoma/genética , Evolução Clonal , Mutação , Metástase Neoplásica
2.
Nanomaterials (Basel) ; 13(9)2023 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-37177046

RESUMO

Recent developments in ultrafine bubble generation have opened up new possibilities for applications in various fields. Herein, we investigated how substances in water affect the size distribution and stability of microbubbles generated by a common nanobubble generator. By combining light scattering techniques with optical microscopy and high-speed imaging, we were able to track the evolution of microbubbles over time during and after bubble generation. Our results showed that air injection generated a higher number of microbubbles (<10 µm) than CO2 injection. Increasing detergent concentration led to a rapid increase in the number of microbubbles generated by both air and CO2 injection and the intensity signal detected by dynamic light scattering (DLS) slightly increased. This suggested that surface-active molecules may inhibit the growth and coalescence of bubbles. In contrast, we found that salts (NaCl and Na2CO3) in water did not significantly affect the number or size distribution of bubbles. Interestingly, the presence of oil in water increased the intensity signal and we observed that the bubbles were coated with an oil layer. This may contribute to the stability of bubbles. Overall, our study sheds light on the effects of common impurities on bubble generation and provides insights for analyzing dispersed bubbles in bulk.

3.
Sci Adv ; 8(7): eabl4923, 2022 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-35171681

RESUMO

Malignant tumors will become vulnerable if their uncontrolled biosynthesis and energy consumption engaged in metabolic reprogramming can be cut off. Here, we report finding a glycolytic inhibitor targeting glioblastoma with graphite dots-assisted laser desorption/ionization mass spectrometry as an integrated drug screening and pharmacokinetic platform (GLMSD). We have performed high-throughput virtual screening to narrow an initial library of 240,000 compounds down to the docking of 40 compounds and identified five previously unknown chemical scaffolds as promising hexokinase-2 inhibitors. The best inhibitor (Compd 27) can regulate the reprogrammed metabolic pathway in U87 glioma cells (median inhibitory concentration ~ 11.3 µM) for tumor suppression. Highly effective therapy against glioblastoma has been demonstrated in both subcutaneous and orthotopic brain tumors by synergizing Compd 27 and temozolomide. Our glycolytic inhibitor discovery can inspire personalized medicine targeting reprogrammed metabolisms of malignant tumors. GLMSD enables large, high-quality data for next-generation artificial intelligence-aided drug development.


Assuntos
Glioblastoma , Grafite , Inteligência Artificial , Linhagem Celular Tumoral , Glioblastoma/metabolismo , Humanos , Espectrometria de Massas , Temozolomida/farmacologia , Temozolomida/uso terapêutico
4.
Antib Ther ; 4(4): 228-241, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34805746

RESUMO

BACKGROUND: Bispecific T cell engaging antibodies (TEAs) with one arm targeting a cancer antigen and another arm binding to CD3 have demonstrated impressive efficacy in multiple clinical studies. However, establishing a safety/efficacy balance remains challenging. For instance, some TEAs have severe safety issues. Additionally, not all patients or all cancer cells of one patient respond equally to TEAs. METHODS: Here, we developed a next-generation bispecific TEA with better safety/efficacy balance and expanded mechanisms of action. Using the computer-aided antibody design strategy, we replaced heavy chain complementarity-determining regions (HCDRs) in one Rituximab arm with HCDRs from a CD3 antibody and generated a novel CD20/CD3 bispecific antibody. RESULTS: After series of computer-aided sequence optimization, the lead molecule, GB261, showed great safety/efficacy balance both in vitro and in animal studies. GB261 exhibited high affinity to CD20 and ultra-low affinity to CD3. It showed comparable T cell activation and reduced cytokine secretion compared with a benchmark antibody (BM). ADCC and CDC caused by GB261 only killed CD20+ cells but not CD3+ cells. It exhibited better RRCL cell killing than the BM in a PBMC-engrafted, therapeutic treatment mouse model and good safety in cynomolgus monkeys. CONCLUSIONS: Thus, GB261 is a promising novel TEA against CD20+ cancers.

5.
J Exp Clin Cancer Res ; 39(1): 273, 2020 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-33278894

RESUMO

BACKGROUND: MUC18 is a glycoprotein highly expressed on the surface of melanoma and other cancers which promotes tumor progression and metastasis. However, its mechanism of action and suitability as a therapeutic target are unknown. METHODS: A monoclonal antibody (mAb) (JM1-24-3) was generated from metastatic melanoma tumor live cell immunization, and high-throughput screening identified MUC18 as the target. RESULTS: Analysis of molecular interactions between MUC18 and JM1-24-3 revealed that the downstream signaling events depended on binding of the mAb to a conformational epitope on the extracellular domain of MUC18. JM1-24-3 inhibited melanoma cell proliferation, migration and invasion in vitro and reduced tumor growth and metastasis in vivo. CONCLUSION: These results confirm that MUC18 is mechanistically important in melanoma growth and metastasis, suggest that the MUC18 epitope identified is a promising therapeutic target, and that the JM1-24-3 mAb may serve as the basis for a potential therapeutic agent.


Assuntos
Anticorpos Monoclonais/farmacologia , Melanoma/terapia , Animais , Anticorpos Monoclonais/imunologia , Antígeno CD146/imunologia , Linhagem Celular Tumoral , Humanos , Masculino , Melanoma/imunologia , Camundongos , Camundongos Endogâmicos A , Camundongos Nus , Distribuição Aleatória , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Sci Rep ; 10(1): 2986, 2020 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-32076029

RESUMO

HER2 overexpression is frequently associated with tumor metastasis and poor prognosis of breast cancer. More evidence indicates that HER3 is involved in HER2-resistant therapies. Combination treatments with two or more different monoclonal antibodies are a promising strategy to overcome resistance to HER2 therapies. We presented a novel fully human HER2-targeted monoclonal antibody, GB235, screened from a phage-display library against the HER2 antigen. GB235 in combination with Trastuzumab overcomes resistance in HER2-positive tumors and results in more sustained inhibition of tumor growth over time. The competition binding assay showed that the epitopes of GB235 do not overlap with those of Pertuzumab and Trastuzumab on HER2. Further HER2 mutagenesis results revealed that the binding epitopes of GB235 were located in the domain III of HER2. The mechanism of action of GB235 in blocking HER2-driven tumors is different from the mechanisms of Trastuzumab or Pertuzumab. GB235 does not affect the heterodimerization of HER2 and HER3, whereas the GB235 combined treatment with Trastuzumab significantly inhibited heregulin-induced HER3 phosphorylation and downstream signaling. Moreover, GB235 in combination with Trastuzumab reversed the resistance to heregulin-induced proliferation in HER2-overexpressing cancer cell lines. GB235 combined with Trastuzumab treatment in xenograft models resulted in improved antitumor activity. Complete tumor suppression was observed in the HER2-positive NCI-N87 xenograft model treated with the combination treatment with GB235 and Trastuzumab. In a Trastuzumab-resistant patient-derived tumor xenograft model GA0060, GB235 plus Trastuzumab reversed the resistance to Trastuzumab monotherapy. Because GB235 showed a different working mechanism with Pertuzumab and Trastuzumab, these agents can be considered complementary therapy against HER2 overexpression tumors.


Assuntos
Anticorpos Monoclonais/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Receptor ErbB-2/antagonistas & inibidores , Animais , Anticorpos Monoclonais/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Neoplasias/patologia , Neuregulina-1/metabolismo , Fosforilação/efeitos dos fármacos , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Trastuzumab/farmacologia , Trastuzumab/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Oncotarget ; 8(19): 31901-31914, 2017 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-28404874

RESUMO

Lysine-specific demethylase 1 (LSD1) has been recognized as a potential therapeutic target for acute myeloid leukemia (AML). Herein, we identified a novel LSD1 inhibitor, JL1037, via Computer Aided Drug Design technology. JL1037 is a potent, selective and reversible LSD1 inhibitor with IC50s of 0.1 µM and >1.5 µM for LSD1 and monoamine oxidases A/B (MAO-A/B), respectively. Treatment of THP-1 and Kasumi-1 cell lines with JL1037 resulted in dose dependent accumulation of H3K4me1 and H3K4me2, the major substrates of LSD1, as well as inhibition of cell proliferation, blockade of cell cycle and induction of apoptosis. Further investigations demonstrated that JL1037 could upregulate cell cycle-related proteins P21, P57, pro-apoptotic protein Bax and downregulate anti-apoptosis proteins Bcl-2 and Bcl-XL. JL1037 appeared to activate autophage response in AML cell lines as well as primary cells from AML patients by increasing LC3-II expression and the formation of autophagosomes and autolysosomes in cytoplasm. Co-treatment with autophagy inhibitor chloroquine (CQ) enhanced JL1037-induced cell apoptosis. Moreover, daily intravenous administration of JL1037 tended to reduce tumor burden and prolong the survival of t(8;21) leukemia mice. In conclusion, JL1037 exhibited potent anti-leukemia effect and could be a potential therapeutic agent for AML treatment.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Histona Desmetilases/antagonistas & inibidores , Animais , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Modelos Animais de Doenças , Ativação Enzimática/efeitos dos fármacos , Humanos , Leucemia Mieloide Aguda/metabolismo , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Clin Invest Med ; 36(3): E133-42, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23739667

RESUMO

PURPOSE: Gold nanoparticles (GNPs) have attracted significant attention in the treatment of cancer due to their potential as novel radiation enhancers, particularly when functionalized with various targeting ligands. The aim of this study was to assess the biodistribution and pharmacokinetic characteristics of a novel choline-bound GNP (choline-GNP) stabilized with polyethelenimine (PEI). METHODS: Choline bound to 27 nm diameter GNPs was characterized using transmission electron microscopy (TEM), X-ray photoelectron spectroscopy (XPS) and Fourier transform infrared spectroscopy (FTIR). Toxicity of choline-GNPs was examined on DU-145 prostate cancer cells using an MTT assay. Using balb/c mice bearing flank DU-145 prostate tumors, choline-GNPs bio-distribution was measured using inductively coupled mass spectroscopy (ICP-MS). Blood, heart, lung, liver, spleen, brain, kidney and tumor gold content were examined at multiple time points over a 24-hour period after tail vein injection. RESULTS: An MTT assay using DU-145 prostate cancer cells yielded a 95% cell viability 72 hours after choline-GNP administration. The tumor GNP area under the concentration-time curve during the first 4 hours (AUC0-4) was 2.2 µg/ml h, representing 13% of the circulating blood GNP concentration over the same time period. The maximum intra-tumor GNP concentration observed was 1.4% of the injected dose per gram of tumor tissue (%ID/g) one hour post injection. CONCLUSIONS: GNPs functionalized with choline demonstrates a viable future nanoparticle platform with increased intra-tumor uptake as compared to unconjugated GNPs. Decreased intra-hepatic accumulation appears to be the reason for the improved systemic bioavailability. The next logical translational investigation will incorporate external beam radiation with the observed maximum intra-tumor uptake.


Assuntos
Colina/metabolismo , Ouro/química , Nanopartículas Metálicas/química , Neoplasias da Próstata/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Masculino , Nanopartículas Metálicas/uso terapêutico , Camundongos , Espectroscopia Fotoeletrônica , Neoplasias da Próstata/metabolismo , Espectroscopia de Infravermelho com Transformada de Fourier
9.
Mol Cancer Ther ; 10(5): 770-83, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21388971

RESUMO

Inhibition of VEGFR-2 signaling reduces angiogenesis and retards tumor growth. Current biotherapeutics that inhibit VEGFR-2 signaling by either sequestering VEGF ligand or inhibiting VEGF binding to VEGFR-2 may be compromised by high VEGF concentrations. Here we describe a biotherapeutic that targets VEGFR-2 signaling by binding to Ig domains 4-7 of VEGFR-2 and therefore has the potential to work independently of ligand concentration. 33C3, a fully human VEGFR-2 antibody, was generated using XenoMouse technology. To elucidate the mechanism of action of 33C3, we have used a number of competition and binding assays. We show that 33C3 binds VEGFR-2 Ig domains 4-7, has no impact on VEGF-A binding to VEGFR-2, and does not compete with an antibody that interacts at the ligand binding site. 33C3 has a high affinity for VEGFR-2 (K(D) < 1 nmol/L) and inhibits VEGF-A induced phosphorylation of VEGFR-2 with an IC(50) of 99 ± 3 ng/mL. In vitro, in a 2D angiogenesis assay, 33C3 potently inhibits both tube length and number of branch points, and endothelial tubule formation in a 3D assay. In vivo, 33C3 is a very effective inhibitor of angiogenesis in both a human endothelial angiogenesis assay and in a human skin chimera model. These data show targeting VEGFR-2 outside of the ligand binding domain results in potent inhibition of VEGFR-2 signaling and inhibition of angiogenesis in vitro and in vivo.


Assuntos
Inibidores da Angiogênese/metabolismo , Anticorpos/metabolismo , Ligantes , Neovascularização Fisiológica/efeitos dos fármacos , Pele/irrigação sanguínea , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Inibidores da Angiogênese/farmacologia , Animais , Anticorpos/farmacologia , Especificidade de Anticorpos/imunologia , Linhagem Celular , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Humanos , Camundongos , Camundongos SCID , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Pele/efeitos dos fármacos , Suínos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/química
10.
J Med Chem ; 54(9): 3368-85, 2011 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-21438541

RESUMO

A novel class of heat shock protein 90 (Hsp90) inhibitors was discovered by high-throughput screening and was subsequently optimized using a combination of structure-based design, parallel synthesis, and the application of medicinal chemistry principles. Through this process, the biochemical and cell-based potency of the original HTS lead were substantially improved along with the corresponding metabolic stability properties. These efforts culminated with the identification of a development candidate (compound 42) which displayed desired PK/PD relationships, significant efficacy in a melanoma A2058 xenograft tumor model, and attractive DMPK profiles.


Assuntos
Antineoplásicos/síntese química , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Pirazóis/síntese química , Pirimidinas/síntese química , Administração Oral , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Ligação Competitiva , Disponibilidade Biológica , Proteínas Sanguíneas/metabolismo , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular , Ensaios de Seleção de Medicamentos Antitumorais , Estabilidade de Medicamentos , Feminino , Humanos , Interações Hidrofóbicas e Hidrofílicas , Técnicas In Vitro , Masculino , Melanoma/tratamento farmacológico , Melanoma/patologia , Camundongos , Camundongos Nus , Microssomos Hepáticos/metabolismo , Modelos Moleculares , Transplante de Neoplasias , Ligação Proteica , Pirazóis/farmacocinética , Pirazóis/farmacologia , Pirimidinas/farmacocinética , Pirimidinas/farmacologia , Ratos , Relação Estrutura-Atividade , Transplante Heterólogo
11.
N Z Dent J ; 106(2): 55-60, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20608309

RESUMO

UNLABELLED: The biocompatibility of metal alloys has generated much concern for practitioners and patients alike over recent years. OBJECTIVES: To investigate dentists' experience of patient allergies to metal alloys used in prosthodontic restorations. DESIGN: Cross-sectional survey of New Zealand practising general dentists. METHODS: A random sample of 700 was taken from the New Zealand dental register. The questionnaire asked dentists whether any of their patients have encountered any allergic reactions to metal alloys. It also sought information on what alloys were being prescribed for use in different types of prosthodontic restorations. RESULTS: A response rate of 71.4% was obtained (N = 476). Some 83 dentists (17.4%) reported having encountered suspected metal allergies in patients; of those, 70 had had the allergies confirmed with a biopsy. Of the entire sample, 327 dentists (72.2%) were aware of the metals used in their restorations, and 201 (44.8%) specified the alloys used in their restorations. For cast removable prosthodontic restorations (such as removable partial dentures), base metal alloys were the most preferred choice; for full cast crowns, high noble alloys were the most favoured; noble alloys were the most favoured for both porcelain-fused-to-metal crowns and fixed-bridge restorations. CONCLUSION: As many as one in six general practising dentists have encountered allergic reactions to metal alloys in their patients. General practising dentists' awareness of the indications for the various metal alloys used in prosthodontic restorations should be raised, and biocompatibility issues should be clarified, so that dentists prescribe the optimum metal alloy for each type of restoration.


Assuntos
Ligas Dentárias , Odontologia Geral/estatística & dados numéricos , Hipersensibilidade/etiologia , Padrões de Prática Odontológica/estatística & dados numéricos , Adulto , Materiais Biocompatíveis , Distribuição de Qui-Quadrado , Estudos Transversais , Ligas Dentárias/efeitos adversos , Prótese Dentária/estatística & dados numéricos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Nova Zelândia , Estudos de Amostragem , Inquéritos e Questionários , Adulto Jovem
12.
Mol Cancer Ther ; 9(1): 145-56, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20053776

RESUMO

Localized angiopoietin-2 (Ang2) expression has been shown to function as a key regulator of blood vessel remodeling and tumor angiogenesis, making it an attractive candidate for antiangiogenic therapy. A fully human monoclonal antibody (3.19.3) was developed, which may have significant pharmaceutical advantages over synthetic peptide-based approaches in terms of reduced immunogenicity and increased half-life to block Ang2 function. The 3.19.3 antibody potently binds Ang2 with an equilibrium dissociation constant of 86 pmol/L, leading to inhibition of Tie2 receptor phosphorylation in cell-based assays. In preclinical models, 3.19.3 treatment blocked blood vessel formation in Matrigel plug assays and in human tumor xenografts. In vivo studies with 3.19.3 consistently showed broad antitumor activity as a single agent across a panel of diverse subcutaneous and orthotopic xenograft models. Combination studies of 3.19.3 with cytotoxic drugs or anti-vascular endothelial growth factor agents showed significant improvements in antitumor activity over single-agent treatments alone with no apparent evidence of increased toxicity. Initial pharmacokinetic profiling studies in mice and nonhuman primates suggested that 3.19.3 has a predicted human half-life of 10 to 14 days. These studies provide preclinical data for 3.19.3 as a potential new antiangiogenic therapy as a single agent or in combination with chemotherapy or vascular endothelial growth factor inhibitors for the treatment of cancer.


Assuntos
Angiopoietina-2/imunologia , Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/uso terapêutico , Especificidade de Anticorpos/efeitos dos fármacos , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Morte Celular/efeitos dos fármacos , Colágeno/metabolismo , Combinação de Medicamentos , Humanos , Laminina/metabolismo , Camundongos , Neovascularização Patológica/tratamento farmacológico , Fosforilação/efeitos dos fármacos , Primatas , Ligação Proteica/efeitos dos fármacos , Proteoglicanas/metabolismo , Receptor TIE-2/metabolismo
13.
Bioorg Med Chem Lett ; 18(23): 6273-8, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18929486

RESUMO

Information from X-ray crystal structures were used to optimize the potency of a HTS hit in a Hsp90 competitive binding assay. A class of novel and potent small molecule Hsp90 inhibitors were thereby identified. Enantio-pure compounds 31 and 33 were potent in PGA-based competitive binding assay and inhibited proliferation of various human cancer cell lines in vitro, with IC(50) values averaging 20 nM.


Assuntos
Amidas/síntese química , Amidas/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Amidas/química , Aminoácidos/química , Antineoplásicos/química , Técnicas de Química Combinatória , Cristalografia por Raios X , Desenho de Fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Chaperonas Moleculares/metabolismo , Conformação Molecular , Estrutura Molecular , Relação Estrutura-Atividade
14.
J Virol ; 78(10): 5438-47, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15113922

RESUMO

Bovine herpesvirus 1 (BHV-1), like other Alphaherpesvirinae subfamily members, establishes latency in sensory neurons. The latency-related (LR) RNA is abundantly expressed during latency, and expression of an LR protein is required for the latency reactivation cycle in cattle. Within LR promoter sequences, a 135-amino-acid open reading frame (ORF) was identified, ORF-E, that is antisense to the LR RNA. ORF-E is also downstream of the gene encoding the major viral transcriptional activator, bICP0. Strand-specific reverse transcription-PCR demonstrated that a transcript containing ORF-E was consistently expressed in trigeminal ganglia (TG) of latently infected calves, productively infected cultured cells, and acutely infected calves. As expected, a late transcript encoding glycoprotein C was not detected in TG of latently infected calves. The ORF-E transcript is polyadenylated and is expressed early when cultured bovine cells are productively infected. Protein coding sequences containing ORF-E were fused to green fluorescent protein (GFP) to examine the cellular localization of the putative protein. In transiently transfected mouse neuroblastoma (neuro-2A) and human neuroblastoma (SK-N-SH) cells, the ORF-E/GFP fusion protein was detected in discreet domains within the nucleus. In contrast, the ORF-E/GFP fusion protein was detected in the cytoplasm and nucleus of rabbit skin cells and bovine kidney cells. As expected, the GFP protein was expressed in the cytoplasm and nucleus of transfected cells. These studies indicate that the ORF-E transcript is consistently expressed during latency. We suggest that the ORF-E gene regulates some aspect of the latency reactivation cycle.


Assuntos
Doenças dos Bovinos/virologia , Infecções por Herpesviridae/veterinária , Herpesvirus Bovino 1/genética , Fases de Leitura Aberta , RNA Mensageiro/análise , RNA Viral/análise , Gânglio Trigeminal/virologia , Latência Viral , Animais , Sequência de Bases , Bovinos , Infecções por Herpesviridae/virologia , Dados de Sequência Molecular , Transfecção
15.
J Pharmacol Exp Ther ; 303(2): 540-8, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12388634

RESUMO

Interferon-alpha (IFN-alpha) is indicated for the treatment of certain viral infections including hepatitis B and C, and cancers such as melanoma. The short circulating half-life of unmodified IFN-alpha makes frequent dosing (daily or three times weekly) over an extended period (6-12 months or more) necessary. To improve the pharmacokinetics of IFN-alpha and decrease dosing frequency, IFN-alpha was fused to human serum albumin producing a new protein, Albuferon. In vitro comparisons of Albuferon and IFN-alpha showed similar antiviral and antiproliferative activities, although Albuferon was less potent on a molar basis than IFN-alpha. Pharmacokinetic and pharmacodynamic properties of the fusion protein were enhanced in monkeys. After a single intravenous injection (30 microg/kg,) clearance was 0.9 ml/h/kg, and the terminal half-life was 68 h. After 30 microg/kg subcutaneous injection, apparent clearance (clearance divided by bioavailability) was 1.4 ml/h/kg, the terminal half-life was 93 h, and bioavailability was 64%. The rate of clearance of Albuferon was approximately 140-fold slower, and the half-life 18-fold longer, than for IFN-alpha given by the subcutaneous route in other monkey studies. Sera from Albuferon-treated monkeys demonstrated dose-related antiviral activity for > or =8 days based on an in vitro bioassay, whereas antiviral activity from IFN-alpha-treated animals was only slightly elevated relative to vehicle on day 0. Significant increases in 2',5'-oligoadenylate synthetase mRNA relative to IFN-alpha- or vehicle-treated animals were maintained for > or =10 days after subcutaneous dosing. The improved pharmacokinetics of Albuferon are accompanied by an improved pharmacodynamic response suggesting that Albuferon may offer the benefits of less frequent dosing and a potentially improved efficacy profile compared with IFN-alpha.


Assuntos
Antivirais/farmacologia , Antivirais/farmacocinética , Interferon-alfa/farmacologia , Interferon-alfa/farmacocinética , 2',5'-Oligoadenilato Sintetase/antagonistas & inibidores , Animais , Antivirais/administração & dosagem , Área Sob a Curva , Disponibilidade Biológica , Linfoma de Burkitt/patologia , Células COS , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Feminino , Humanos , Injeções Intravenosas , Injeções Subcutâneas , Interferon-alfa/administração & dosagem , Interferon-alfa/imunologia , Macaca fascicularis , Masculino , RNA Mensageiro/biossíntese , Albumina Sérica , Albumina Sérica Humana
16.
Immunity ; 16(3): 479-92, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11911831

RESUMO

DR3 is a death domain-containing receptor that is upregulated during T cell activation and whose overexpression induces apoptosis and NF-kappaB activation in cell lines. Here we show that an endothelial cell-derived TNF-like factor, TL1A, is a ligand for DR3 and decoy receptor TR6/DcR3 and that its expression is inducible by TNF and IL-1alpha. TL1A induces NF-kappaB activation and apoptosis in DR3-expressing cell lines, while TR6-Fc protein antagonizes these signaling events. Interestingly, in T cells, TL1A acts as a costimulator that increases IL-2 responsiveness and secretion of proinflammatory cytokines both in vitro and in vivo. Our data suggest that interaction of TL1A with DR3 promotes T cell expansion during an immune response, whereas TR6 has an opposing effect.


Assuntos
Glicoproteínas de Membrana , Receptores de Superfície Celular/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/genética , Sequência de Aminoácidos , Animais , Humanos , Interleucina-1/genética , Interleucina-1/metabolismo , Ligantes , Ativação Linfocitária/imunologia , Camundongos , Dados de Sequência Molecular , Receptores de Superfície Celular/imunologia , Receptores do Fator de Necrose Tumoral/imunologia , Membro 25 de Receptores de Fatores de Necrose Tumoral , Membro 6b de Receptores do Fator de Necrose Tumoral , Alinhamento de Sequência , Linfócitos T/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA