Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Virol ; 82(14): 6927-34, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18480438

RESUMO

Nearly a third of the human population is at risk of infection with the four serotypes of dengue viruses, and it is estimated that more than 100 million infections occur each year. A licensed vaccine for dengue viruses has become a global health priority. A major challenge to developing a dengue vaccine is the necessity to produce fairly uniform protective immune responses to all four dengue virus serotypes. We have developed two bivalent dengue virus vaccines, using a complex adenovirus vector, by incorporating the genes expressing premembrane (prM) and envelope (E) proteins of dengue virus types 1 and 2 (dengue-1 and -2, respectively) (CAdVax-Den12) or dengue-3 and -4 (CAdVax-Den34). Rhesus macaques were vaccinated by intramuscular inoculation of a tetravalent dengue vaccine formulated by combining the two bivalent vaccine constructs. Vaccinated animals produced high-titer antibodies that neutralized all four serotypes of dengue viruses in vitro. The ability of the vaccine to induce rapid, as well as sustained, protective immune responses was examined with two separate live-virus challenges administered at 4 and 24 weeks after the final vaccination. For both of these virus challenge studies, significant protection from viremia was demonstrated for all four dengue virus serotypes in vaccinated animals. Viremia from dengue-1 and dengue-3 challenges was completely blocked, whereas viremia from dengue-2 and dengue-4 was significantly reduced, as well as delayed, compared to that of control-vaccinated animals. These results demonstrate that the tetravalent dengue vaccine formulation provides significant protection in rhesus macaques against challenge with all four dengue virus serotypes.


Assuntos
Adenoviridae/genética , Vacinas contra Dengue/genética , Vacinas contra Dengue/imunologia , Vírus da Dengue/genética , Vírus da Dengue/imunologia , Dengue/prevenção & controle , Vetores Genéticos , Animais , Anticorpos Antivirais/sangue , Dengue/imunologia , Injeções Intramusculares , Macaca mulatta , Testes de Neutralização , Proteínas Estruturais Virais/genética , Viremia/prevenção & controle
2.
Front Biosci ; 13: 2293-8, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17981711

RESUMO

During the last decade, sphingolipid deregulation, namely the balance between the pro-apoptotic molecule ceramide and the anti-apoptotic sphingolipid sphingosine-1-phosphate, has emerged as an important factor in cancer pathology and resistance to therapy. Thus, our research has been focused on developing drugs that are able to restore normal sphingolipid balance, precisely through increasing the levels of ceramide and decreasing sphingosine-1-phosphate. Particularly, inhibition of the ceramide metabolizing enzyme acid ceramidase, whose over-expression in cancer cells has been implicated in resistance to treatment, is proving to be an efficient and promising strategy. In this review, we consider our recent work with acid ceramidase inhibitors, in combination with radiation or gene therapy as a sensitizer that enhance cancer therapy.


Assuntos
Inibidores Enzimáticos/farmacologia , Galactosilgalactosilglucosilceramidase/antagonistas & inibidores , Terapia Genética/métodos , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Neoplasias/terapia , Animais , Antineoplásicos/uso terapêutico , Vírus da Anemia da Galinha/genética , Proteína Ligante Fas/metabolismo , Vetores Genéticos/metabolismo , Humanos , Neoplasias/metabolismo
3.
Cancer Chemother Pharmacol ; 61(2): 231-42, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17429631

RESUMO

PURPOSE: Alterations in ceramide metabolism have been reported in prostate cancer (PCa), resulting in escape of cancer cells from ceramide-induced apoptosis. Specifically, increased expression of lysosomal acid ceramidase (AC) has been shown in some primary PCa tissues and in several PCa cell lines. To determine if this represents a novel therapeutic target, we designed and synthesized LCL204, a lysosomotropic analog of B13, a previously reported inhibitor of AC METHODS: Prostate cancer cell lines were treated with LCL204 for varying times and concentrations. Effects of treatment on cytotoxicity, sphingolipid content, and apoptotic markers were assessed. RESULTS: Treatment of DU145 PCa cells resulted in increased ceramide and decreased sphingosine levels. Interestingly, LCL204 caused degradation of AC in a cathepsin-dependent manner. We also observed rapid destabilization of lysosomes and the release of lysosomal proteases into the cytosol following treatment with LCL204. Combined, these events resulted in mitochondria depolarization and executioner caspase activation, ultimately ending in apoptosis CONCLUSIONS: These results provide evidence that treatment with molecules such as LCL204, which restore ceramide levels in PCa cells may serve as a new viable treatment option for PCa.


Assuntos
Antineoplásicos , Inibidores Enzimáticos/farmacologia , Galactosilgalactosilglucosilceramidase/antagonistas & inibidores , Neoplasias da Próstata/tratamento farmacológico , Apoptose/efeitos dos fármacos , Western Blotting , Caspases/metabolismo , Linhagem Celular Tumoral , Ceramidas/metabolismo , Ativação Enzimática/efeitos dos fármacos , Humanos , Concentração de Íons de Hidrogênio , Indicadores e Reagentes , Lisossomos/efeitos dos fármacos , Lisossomos/enzimologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Microscopia Confocal , Membranas Mitocondriais/efeitos dos fármacos , Neoplasias da Próstata/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Esfingolipídeos/metabolismo , Esfingomielina Fosfodiesterase/antagonistas & inibidores , Frações Subcelulares/metabolismo
4.
Mol Ther ; 15(7): 1259-63, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17426710

RESUMO

Head and neck squamous cell cancers (HNSCC) are particularly aggressive and are resistant to many forms of treatment. Ceramide metabolism has been shown to play an important role in cancer progression and cancer resistance to therapy in many tumor models, including HNSCC. Here, we study the role of the ceramide-metabolizing enzyme acid ceramidase (AC) in therapeutic responses in HNSCC. First, we show that AC is over-expressed in 70% of head and neck squamous cell tumors compared with normal tissues, suggesting that this enzyme may play an important role in facilitating HNSCC growth. Next, comparison of three HNSCC cell lines with low, medium, and high levels of AC reveals an inverse correlation between the levels of AC and their response to exogenous C-6-ceramide. Furthermore, over-expression of AC in SCC-1 cells increased resistance to Fas-induced cell killing. Conversely, down-regulation of AC using specific AC small interfering RNA (siRNA) sensitized the SCC-1 cancer cell line to Fas-induced apoptosis. Finally, we show that the AC inhibitor LCL 204 can sensitize HNSCC cell lines to Fas-induced apoptosis both in vitro and in a xenograft model in vivo, suggesting that the combination of FasL gene therapy and LCL 204 may become a new treatment option for advanced-stage head and neck cancer.


Assuntos
Inibidores Enzimáticos/farmacologia , Proteína Ligante Fas/genética , Proteína Ligante Fas/metabolismo , Galactosilgalactosilglucosilceramidase/antagonistas & inibidores , Galactosilgalactosilglucosilceramidase/metabolismo , Terapia Genética , Neoplasias de Cabeça e Pescoço/metabolismo , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Ceramidas/metabolismo , Regulação para Baixo , Feminino , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/genética , Humanos , Camundongos , Camundongos Nus , RNA Interferente Pequeno/genética , Sensibilidade e Especificidade , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Am J Trop Med Hyg ; 76(4): 743-51, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17426182

RESUMO

There are approximately 100 million new cases of dengue (DEN) virus infection each year. Infection can result in illness ranging from a mild fever to hemorrhaging, shock, or even death. There are four serotypes of dengue virus (DEN1-4), and immunity to one serotype does not cross protect from infection with other serotypes. Currently there are no approved vaccines for dengue fever. In this report, we describe the construction of a bivalent dengue virus vaccine using a complex recombinant adenovirus approach to express multiple genes of DEN1 and DEN2 serotypes. In vaccinated mice, this vector induced humoral immune responses against all four dengue serotypes as measured by enzyme-linked immunosorbent assay. However, the neutralizing antibody responses were specific for DEN1 and DEN2 serotypes. Expansion of this vaccine development platform towards the DEN3 and DEN4 serotypes can lead towards the development of an adenovirus-based tetravalent dengue vaccine.


Assuntos
Adenoviridae/genética , Antígenos Virais/genética , Antígenos Virais/imunologia , Vírus da Dengue/genética , Vírus da Dengue/imunologia , Dengue/imunologia , Dengue/virologia , Vacinas Virais/genética , Vacinas Virais/imunologia , Animais , Linhagem Celular , Chlorocebus aethiops , Expressão Gênica , Humanos , Camundongos , Células Vero
6.
FEBS Lett ; 580(19): 4751-6, 2006 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-16901483

RESUMO

Treatment of different cancer cell lines with desipramine induced a time- and dose-dependent downregulation of acid ceramidase. Desipramine's effect on acid ceramidase appeared specific for amphiphilic agents (desipramine, chlorpromazine, and chloroquine) but not other lysomotropic agents such as ammonium chloride and bafilomycin A1, and was not transcriptionally regulated. The cathepsin B/L inhibitor, CA074ME, but not the cathepsin D inhibitor, pepstatin A, blocked desipramine's effect on acid ceramidase. Desipramine led to a more pronounced downregulation of sphingosine compared to ceramide suggesting acid ceramidase inhibition is important to desipramine's mechanism of action. This study reveals a new mechanism of action for desipramine.


Assuntos
Antidepressivos Tricíclicos/farmacologia , Cisteína Endopeptidases/metabolismo , Desipramina/farmacologia , Inibidores Enzimáticos/farmacologia , Galactosilgalactosilglucosilceramidase/antagonistas & inibidores , Linhagem Celular Tumoral , Humanos , Hidrólise , Masculino , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Esfingolipídeos/metabolismo
7.
Cancer Gene Ther ; 12(1): 12-8, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15514684

RESUMO

Previous investigations have revealed that bladder cancer cells are generally resistant to Fas-mediated apoptosis by conventional Fas agonists. However, the ability of these cell lines to undergo Fas-mediated apoptosis may have been underappreciated. As a result, we investigated the in vitro efficacy of Fas ligand gene therapy for bladder cancer. Three human bladder cancer lines (T24, J82, and 5637) were treated with the conventional Fas agonist CH-11, a monoclonal antibody to the Fas receptor. Cells were also treated with a replication-deficient adenovirus containing a modified murine Fas ligand gene fused to green fluorescent protein (GFP), AdGFPFasL. A virus containing the GFP gene alone was used to control for viral toxicity (AdGFP). Cell death was quantified using a tetrazolium-based (MTS) assay. Cells were also evaluated by Western blotting to evaluate poly (ADP-ribose) polymerase, caspase 8, and caspase 9 cleavage and by flow cytometry to determine the presence of coxsackie/adenovirus receptor (CAR). These studies confirmed bladder cancer resistance to cell death by the anti-Fas monoclonal antibody CH-11. This resistance was overcome with AdGFPFasL at a multiplicity of infection (MOI) of 1000 achieving over 80% cell death in all cell lines. Furthermore, greater than 80% cell death was evident in 5637 cells treated with low-dose AdGFPFasL (MOI=10). 5637 cells expressed significantly higher levels of surface CAR than J82 or T24 cells (P<.05). AdGFPFasL is cytotoxic to bladder cancer cells that would otherwise be considered Fas resistant, supporting its in vivo potential. Enhanced sensitivity to AdGFPFasL may be in part due to increased cell surface CAR levels.


Assuntos
Apoptose/genética , Terapia Genética , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/terapia , Adenoviridae , Anticorpos/farmacologia , Anticorpos Monoclonais , Antígenos de Superfície , Proteínas de Ligação ao Cálcio , Proteínas do Olho , Proteína Ligante Fas , Vetores Genéticos , Humanos , Ligantes , Lipoproteínas , Glicoproteínas de Membrana , Recoverina , Células Tumorais Cultivadas , Receptor fas
8.
Clin Vaccine Immunol ; 17(4): 572-81, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20181765

RESUMO

Ebola viruses are highly pathogenic viruses that cause outbreaks of hemorrhagic fever in humans and other primates. To meet the need for a vaccine against the several types of Ebola viruses that cause human diseases, we developed a multivalent vaccine candidate (EBO7) that expresses the glycoproteins of Zaire ebolavirus (ZEBOV) and Sudan ebolavirus (SEBOV) in a single complex adenovirus-based vector (CAdVax). We evaluated our vaccine in nonhuman primates against the parenteral and aerosol routes of lethal challenge. EBO7 vaccine provided protection against both Ebola viruses by either route of infection. Significantly, protection against SEBOV given as an aerosol challenge, which has not previously been shown, could be achieved with a boosting vaccination. These results demonstrate the feasibility of creating a robust, multivalent Ebola virus vaccine that would be effective in the event of a natural virus outbreak or biological threat.


Assuntos
Adenoviridae/genética , Vacinas contra Ebola/imunologia , Ebolavirus/imunologia , Vetores Genéticos , Doença pelo Vírus Ebola/prevenção & controle , Animais , Modelos Animais de Doenças , Vacinas contra Ebola/genética , Ebolavirus/genética , Doença pelo Vírus Ebola/imunologia , Humanos , Imunização Secundária/métodos , Macaca fascicularis , Macaca mulatta , Análise de Sobrevida
9.
Clin Vaccine Immunol ; 16(11): 1624-32, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19776190

RESUMO

Rift Valley fever virus (RVFV) has been cited as a potential biological-weapon threat due to the serious and fatal disease it causes in humans and animals and the fact that this mosquito-borne virus can be lethal in an aerosolized form. Current human and veterinary vaccines against RVFV, however, are outdated, inefficient, and unsafe. We have incorporated the RVFV glycoprotein genes into a nonreplicating complex adenovirus (CAdVax) vector platform to develop a novel RVFV vaccine. Mice vaccinated with the CAdVax-based vaccine produced potent humoral immune responses and were protected against lethal RVFV infection. Additionally, protection was elicited in mice despite preexisting immunity to the adenovirus vector.


Assuntos
Adenoviridae/genética , Vetores Genéticos , Febre do Vale de Rift/prevenção & controle , Vírus da Febre do Vale do Rift/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Masculino , Camundongos , Febre do Vale de Rift/imunologia , Vírus da Febre do Vale do Rift/genética , Análise de Sobrevida , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/imunologia , Vacinas Virais/genética
10.
Vaccine ; 26(21): 2627-39, 2008 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-18395306

RESUMO

There are legitimate concerns that the highly pathogenic H5N1 avian influenza virus could adapt for human-to-human transmission and cause a pandemic similar to the 1918 "Spanish flu" that killed 50 million people worldwide. We have developed pandemic influenza vaccines by incorporating multiple antigens from both avian and Spanish influenza viruses into complex recombinant adenovirus vectors. In vaccinated mice, these vaccines induced strong humoral and cellular immune responses against pandemic influenza virus antigens, and protected vaccinated mice against lethal H5N1 virus challenge. These results indicate that this multi-antigen, broadly protective vaccine may serve as a safer and more effective approach than traditional methods for development of a pandemic influenza vaccine.


Assuntos
Antígenos Virais/imunologia , Vetores Genéticos , Virus da Influenza A Subtipo H5N1/imunologia , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Adenoviridae/genética , Animais , Anticorpos Antivirais/sangue , Peso Corporal , Masculino , Camundongos , Análise de Sobrevida , Linfócitos T/imunologia
11.
Clin Vaccine Immunol ; 15(3): 460-7, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18216185

RESUMO

Filoviruses (Ebola and Marburg viruses) are among the deadliest viruses known to mankind, with mortality rates nearing 90%. These pathogens are highly infectious through contact with infected body fluids and can be easily aerosolized. Additionally, there are currently no licensed vaccines available to prevent filovirus outbreaks. Their high mortality rates and infectious capabilities when aerosolized and the lack of licensed vaccines available to prevent such infectious make Ebola and Marburg viruses serious bioterrorism threats, placing them both on the category A list of bioterrorism agents. Here we describe a panfilovirus vaccine based on a complex adenovirus (CAdVax) technology that expresses multiple antigens from five different filoviruses de novo. Vaccination of nonhuman primates demonstrated 100% protection against infection by two species of Ebola virus and three Marburg virus subtypes, each administered at 1,000 times the lethal dose. This study indicates the feasibility of vaccination against all current filovirus threats in the event of natural hemorrhagic fever outbreak or biological attack.


Assuntos
Adenoviridae/genética , Vacinas contra Ebola , Filoviridae , Vetores Genéticos , Doença pelo Vírus Ebola/prevenção & controle , Doença do Vírus de Marburg/prevenção & controle , Vacinas Virais , Adenoviridae/imunologia , Animais , Antígenos Virais/genética , Antígenos Virais/metabolismo , Bioterrorismo/prevenção & controle , Vacinas contra Ebola/administração & dosagem , Vacinas contra Ebola/genética , Vacinas contra Ebola/imunologia , Ebolavirus/imunologia , Ebolavirus/patogenicidade , Filoviridae/classificação , Filoviridae/genética , Filoviridae/imunologia , Doença pelo Vírus Ebola/imunologia , Humanos , Macaca fascicularis , Doença do Vírus de Marburg/imunologia , Marburgvirus/classificação , Marburgvirus/imunologia , Marburgvirus/patogenicidade , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Vacinas Virais/imunologia
12.
Clin Vaccine Immunol ; 14(2): 182-9, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17192403

RESUMO

Dengue virus infections can cause hemorrhagic fever, shock, encephalitis, and even death. Worldwide, approximately 2.5 billion people live in dengue-infested regions with about 100 million new cases each year, although many of these infections are believed to be silent. There are four antigenically distinct serotypes of dengue virus; thus, immunity from one serotype will not cross-protect from infection with the other three. The difficulties that hamper vaccine development include requirements of the natural conformation of the envelope glycoprotein to induce neutralizing immune responses and the necessity of presenting antigens of all four serotypes. Currently, the only way to meet these requirements is to use a mixture of four serotypes of live attenuated dengue viruses, but safety remains a major problem. In this study, we have developed the basis for a tetravalent dengue vaccine using a novel complex adenovirus platform that is capable of expressing multiple antigens de novo. This dengue vaccine is constructed as a pair of vectors that each expresses the premembrane and envelope genes of two different dengue virus serotypes. Upon vaccination, the vaccine expressed high levels of the dengue virus antigens in cells to mimic a natural infection and induced both humoral and cellular immune responses against multiple serotypes of dengue virus in an animal model. Further analyses show the humoral responses were indeed neutralizing against all four serotypes. Our studies demonstrate the concept of mimicking infections to induce immune responses by synthesizing dengue virus membrane antigens de novo and the feasibility of developing an effective tetravalent dengue vaccine by vector-mediated expression of glycoproteins of the four serotypes.


Assuntos
Adenoviridae , Vacinas contra Dengue/imunologia , Vírus da Dengue/imunologia , Vetores Genéticos , Animais , Linhagem Celular , Chlorocebus aethiops , Dengue/imunologia , Dengue/prevenção & controle , Vírus da Dengue/classificação , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Sorotipagem , Células Vero
13.
Clin Vaccine Immunol ; 14(9): 1117-26, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17634508

RESUMO

West Nile Virus (WNV), a member of the family Flaviviridae, was first identified in Africa in 1937. In recent years, it has spread into Europe and North America. The clinical manifestations of WNV infection range from mild febrile symptoms to fatal encephalitis. Two genetic lineages (lineages I and II) are recognized; lineage II is associated with mild disease, while lineage I has been associated with severe disease, including encephalitis. WNV has now spread across North America, significantly affecting both public and veterinary health. In the efforts to develop an effective vaccine against all genetic variants of WNV, we have studied the feasibility of inducing both neutralizing and cellular immune responses by de novo synthesis of WNV antigens using a complex adenoviral vaccine (CAdVax) vector. By expressing multiple WNV proteins from a single vaccine vector, we were able to induce both humoral and cellular immune responses in vaccinated mice. Neutralization assays demonstrated that the antibodies were broadly neutralizing against both lineages of WNV, with a significant preference for the homologous lineage II virus. The results from this study show that multiple antigens synthesized de novo from a CAdVax vector are capable of inducing both humoral and cellular immune responses against WNV and that a multiantigen approach may provide broad protection against multiple genetic variants of WNV.


Assuntos
Proteínas Virais/imunologia , Febre do Nilo Ocidental/imunologia , Vacinas contra o Vírus do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/imunologia , Adenoviridae/genética , Adenoviridae/imunologia , Animais , Formação de Anticorpos/imunologia , Proteínas do Capsídeo/biossíntese , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/imunologia , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Humanos , Imunidade Celular/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Células Vero , Proteínas do Envelope Viral/biossíntese , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Proteínas não Estruturais Virais/biossíntese , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia , Proteínas Virais/biossíntese , Proteínas Virais/genética , Febre do Nilo Ocidental/prevenção & controle , Vacinas contra o Vírus do Nilo Ocidental/genética
14.
Vaccine ; 24(15): 2975-86, 2006 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-16530297

RESUMO

Marburg virus (MARV) is an African filovirus that causes a deadly hemorrhagic fever in humans, with up to 90% mortality. Currently, there are no MARV vaccines or therapies approved for human use. We hypothesized that developing a vaccine that induces a de novo synthesis of MARV antigens in vivo will lead to strong induction of both a humoral and cell-mediated immune response against MARV. Here, we develop and characterize three novel gene-based vaccine candidates which express the viral glycoprotein (GP) from either the Ci67, Ravn or Musoke strain of MARV. Immunization of mice with complex adenovirus (Ad)-based vaccine candidates (cAdVax vaccines), led to efficient production of both antibodies and cytotoxic T lymphocytes (CTL) specific to Musoke strain GP and Ci67 strain GP, respectively. Antibody responses were also shown to be cross-reactive across the MARV strains, but not cross-reactive to Ebola virus, a related filovirus. Additionally, three 1 x 10(8)pfu doses of vaccine vector were demonstrated to be safe in mice, as this did not lead to any detectable toxicity in liver or spleen. These promising results indicate that a cAdVax-based vaccine could be effective for induction of both humoral and cell-mediated immune responses to multiple strains of the Marburg virus.


Assuntos
Adenoviridae/genética , Doença do Vírus de Marburg/prevenção & controle , Marburgvirus/genética , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Antígenos Virais/biossíntese , Antígenos Virais/genética , Antígenos Virais/imunologia , Reações Cruzadas , Testes Imunológicos de Citotoxicidade , Ensaio de Imunoadsorção Enzimática , Vetores Genéticos , Humanos , Fígado/patologia , Doença do Vírus de Marburg/imunologia , Marburgvirus/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Baço/patologia , Linfócitos T Citotóxicos/imunologia , Proteínas do Envelope Viral/biossíntese , Proteínas do Envelope Viral/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/efeitos adversos , Vacinas Virais/genética
15.
Mol Ther ; 14(5): 627-36, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16926120

RESUMO

The potential anti-tumor agent Apoptin activates apoptosis in many human cancers and transformed cell lines, but is believed to be less potent in primary cells. Although caspase 3 is activated during apoptin-induced apoptosis, the mechanism of tumor cell killing remains elusive. We now show that apoptin-mediated cell death involves modulation of the sphingomyelin-ceramide pathway. Treating cells with Ad-GFPApoptin resulted in increased ceramide accumulation and enhanced expression of acid sphingomyelinase (ASMase) with a concomitant increase in ASMase activity and decreased sphingomyelin. Using confocal microscopy, ASMase, normally present in the endosomal/lysosomal compartment, was observed to translocate to the cell's periphery. Cotreatment of Ad-GFPApoptin-infected cells with the ASMase inhibitor desipramine (2.5 muM) attenuated (30%; P<0.01) apoptin-induced cell death. Apoptin was also able to induce a significant decline in sphingosine content by inhibition of ceramide deacylation through down-regulation of acid ceramidase at the protein level. Supporting the role of ceramide in apoptin action, treatment of cells with the combination of an exogenous cell-permeable ceramide analog (C6-ceramide) and Ad-GFPApoptin infection yielded a significant increase (P<0.01) in apoptosis over either treatment modality alone. Together, these data suggest that apoptin modulates ceramide/sphingolipid metabolism as part of its mechanism of action.


Assuntos
Apoptose , Proteínas do Capsídeo/metabolismo , Esfingolipídeos/metabolismo , Adenoviridae/genética , Apoptose/efeitos dos fármacos , Proteínas do Capsídeo/genética , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Ceramidas/biossíntese , Desipramina/farmacologia , Regulação para Baixo , Endossomos/metabolismo , Galactosilgalactosilglucosilceramidase/metabolismo , Expressão Gênica , Genes Reporter/genética , Humanos , Lisossomos/metabolismo , Masculino , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Transporte Proteico , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Esfingomielina Fosfodiesterase/metabolismo , Regulação para Cima
16.
Future Oncol ; 1(1): 115-23, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16555981

RESUMO

Gene therapy has been in a continuous evolutionary process since the first approved trial occurred in 1990 at the National Institute of Health. In the USA, as of March 2004, there were 619 approved gene therapy/transfer protocols and 405 of these were for cancer treatment. Another 294 trials are in progress worldwide, with most concentrated in Europe. However, cancer gene therapy is in its relative infancy when compared with the well-established use of chemo-radiotherapy for treating cancer. As the field develops it is becoming clear that using gene therapy in conjunction with established chemo-radiotherapy approaches is yielding the best results. This concept shall be reviewed in the context of the status of the field, and a future direction based on a combination of gene therapy with small molecule modification of sphingolipid metabolism shall be discussed.


Assuntos
Adenoviridae , Carcinoma de Células Escamosas/terapia , Terapia Genética , Neoplasias de Cabeça e Pescoço/terapia , Neoplasias da Próstata/terapia , Amidoidrolases/antagonistas & inibidores , Ceramidases , Ceramidas/metabolismo , Citosina Desaminase/genética , Proteína Ligante Fas , Genes p53 , Humanos , Masculino , Glicoproteínas de Membrana/genética , Timidina Quinase/genética , Fatores de Necrose Tumoral/genética , Vacinas Virais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA