Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Front Neurosci ; 15: 596976, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34149342

RESUMO

The blood-brain barrier (BBB) hinders the distribution of therapeutics intended for treatment of diseases of the brain. Our previous studies demonstrated that that a soluble form of melanotransferrin (MTf; Uniprot P08582; also known as p97, MFI2, and CD228), a mammalian iron-transport protein, is an effective carrier for delivery of drug conjugates across the BBB into the brain and was the first BBB targeting delivery system to demonstrate therapeutic efficacy within the brain. Here, we performed a screen to identify peptides from MTf capable of traversing the BBB. We identified a highly conserved 12-amino acid peptide, termed MTfp, that retains the ability to cross the intact BBB intact, distributes throughout the parenchyma, and enter endosomes and lysosomes within neurons, astrocytes and microglia in the brain. This peptide may provide a platform for the transport of therapeutics to the CNS, and thereby offers new avenues for potential treatments of neuropathologies that are currently refractory to existing therapies.

2.
Front Mol Biosci ; 8: 611367, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33869275

RESUMO

The blood-brain barrier (BBB) hinders the distribution of therapeutics intended for treatment of neuroinflammation (NI) of the central nervous system. A twelve-amino acid peptide that transcytoses the BBB, termed MTfp, was chemically conjugated to siRNA to create a novel peptide-oligonucleotide conjugate (POC), directed to downregulate NOX4, a gene thought responsible for oxidative stress in ischemic stroke. The MTfp-NOX4 POC has the ability to cross the intact BBB and knockdown NOX4 expression in the brain. Following induction of ischemic stroke, animals pretreated with the POC exhibited significantly smaller infarcts; accompanied by increased protection against neurological deterioration and improved recovery. The data demonstrates that the MTfp can act as a nanomule to facilitate BBB transcytosis of siRNAs; where the NOX-4 specific siRNA moiety can elicit effective therapeutic knockdown of a gene responsible for oxidative stress in the central nervous system. This study is the first to conclusively demonstrate both siRNA-carrier delivery and therapeutic efficacy in any CNS disease model where the BBB remains intact and thus offers new avenues for potential treatments of oxidative stress underlying neuroinflammation in a variety of neuropathologies that are currently refractory to existing therapies.

4.
Clin Cancer Res ; 14(5): 1494-501, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18316574

RESUMO

PURPOSE: Tpn is a member of the MHC class I loading complex and functions to bridge the TAP peptide transporter to MHC class I molecules. Metastatic human carcinomas often express low levels of the antigen-processing components Tapasin and TAP and display few functional surface MHC class I molecules. As a result, carcinomas are unrecognizable by effector CTLs. The aim of this study is to examine if Tapasin (Tpn) plays a critical role in the escape of tumors from immunologic recognition. EXPERIMENTAL DESIGN: To test our hypothesis, a nonreplicating adenovirus vector encoding human Tpn (AdhTpn) was constructed to restore Tpn expression in vitro and in vivo in a murine lung carcinoma cell line (CMT.64) that is characterized by down-regulation of surface MHC class I due to deficiency in antigen-processing components. RESULTS: Ex vivo, Tpn expression increased surface MHC class I and restored susceptibility of tumor cells to antigen-specific CTL killing, and AdhTpn infection of dendritic cells also significantly increased cross-presentation and cross-priming. Furthermore, tumor-bearing animals inoculated with AdhTpn demonstrated a significant increase in CD8(+) and CD4(+) T cells and CD11c(+) dendritic cells infiltrating the tumors. Provocatively, whereas syngeneic mice bearing tumors that were inoculated with AdhTpn a significant reduction in tumor growth and increased survival compared with vector controls, combining AdhTpn inoculation with AdhTAP1 resulted in a significant augmentation of protection from tumor-induced death than either component alone. CONCLUSIONS: This is the first demonstration that Tpn alone can enhance survival and immunity against tumors but additionally suggests that Tpn and TAP should be used together as components of immunotherapeutic vaccine protocols to eradicate tumors.


Assuntos
Transportadores de Cassetes de Ligação de ATP/imunologia , Apresentação de Antígeno , Neoplasias Pulmonares/terapia , Proteínas de Membrana Transportadoras/imunologia , Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Adenoviridae/genética , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Apresentação Cruzada , Células Dendríticas/imunologia , Intervalo Livre de Doença , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Neoplasias Pulmonares/imunologia , Linfócitos do Interstício Tumoral , Camundongos , Camundongos Endogâmicos C57BL , Baço/citologia , Baço/imunologia , Baço/metabolismo , Taxa de Sobrevida , Linfócitos T Citotóxicos
5.
J Cereb Blood Flow Metab ; 39(10): 2074-2088, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-29845881

RESUMO

Delivery of biologic drugs across the blood-brain barrier is becoming a reality. However, the solutions often involve the assembly of complex multi-specific antibody molecules. Here we utilize a simple 12 amino-acid peptide originating from the melanotransferrin (MTf) protein that has shown improved brain delivery properties. 3D confocal fluorescence microscopic analysis demonstrated brain parenchymal localisation of a fluorescently labelled antibody (NIP228) when chemically conjugated to either the MTf peptide or full-length MTf protein. Measurement of plasma kinetics demonstrated the MTf peptide fusions had very similar kinetics to an unmodified NIP228 control antibody, whereas the fusion to MTf protein had significantly reduced plasma exposure most likely due to a higher tissue distribution in the periphery. Brain exposure for the MTf peptide fusions was significantly increased for the duration of the study, exceeding that of the fusions to full length MTf protein. Using a neuropathic pain model, we have demonstrated that fusions to interleukin-1 receptor antagonist (IL-1RA) are able to induce significant and durable analgesia following peripheral administration. These data demonstrate that recombinant and chemically conjugated MTf-based brain delivery vectors can deliver therapeutic levels of drug to the central nervous system.


Assuntos
Portadores de Fármacos/metabolismo , Proteína Antagonista do Receptor de Interleucina 1/uso terapêutico , Glicoproteínas de Membrana/metabolismo , Neuralgia/tratamento farmacológico , Peptídeos/metabolismo , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Portadores de Fármacos/química , Humanos , Proteína Antagonista do Receptor de Interleucina 1/administração & dosagem , Proteína Antagonista do Receptor de Interleucina 1/farmacocinética , Masculino , Glicoproteínas de Membrana/química , Camundongos Endogâmicos C57BL , Neuralgia/metabolismo , Peptídeos/química
6.
Cancer Res ; 65(17): 7926-33, 2005 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16140964

RESUMO

A wide variety of human carcinomas have low expression of tumor-associated antigen presentation in the context of MHC class I antigens due to defects in the antigen presentation pathway. This immune evasion mechanism renders many tumors unrecognizable by host immune surveillance mechanisms. The present study examines the expression of HLA, tapasin, transporter associated with antigen processing 1 (TAP1), and beta2 microglobulin in human small cell lung carcinoma and non-small cell lung carcinoma. Immunohistochemical staining showed severe impairment of the antigen presentation pathway in all patients. In order to recover tumor immunogenicity, a nonreplicating adenovirus expressing human TAP1 (AdhTAP1) was used to restore the expression of TAP1 in the antigen presentation pathway-deficient mouse lung carcinoma cell line, CMT.64. Infection of CMT.64 cells with AdhTAP1 increased MHC class I antigen surface expression, antigen presentation, and susceptibility to antigen-specific CTLs. Fluorescence-activated cell sorting and ELISPOT analysis showed that AdhTAP1 treatment significantly increased dendritic cell cross-presentation and cross-priming of tumor antigens. Furthermore, ex vivo and in vivo AdhTAP1 treatment significantly retarded tumor growth and increased survival of mice bearing CMT.64 tumors. Fluorescence-activated cell sorting analysis and immunohistochemical staining showed a significant increase in CD8+ and CD4+ T cells and CD11c+ dendritic cells infiltrating the tumors. The results show that TAP should be considered as a part of the immunotherapies for various cancers because it is likely to provide a general method for increasing immune responses against tumors regardless of the antigenic composition of the tumor or the MHC haplotypes of the host.


Assuntos
Transportadores de Cassetes de Ligação de ATP/biossíntese , Apresentação de Antígeno/imunologia , Antiporters/biossíntese , Antígenos HLA/biossíntese , Imunoglobulinas/biossíntese , Neoplasias Pulmonares/imunologia , Microglobulina beta-2/biossíntese , Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/imunologia , Adenoviridae/genética , Idoso , Antiporters/genética , Antiporters/imunologia , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma de Células Pequenas/genética , Carcinoma de Células Pequenas/imunologia , Carcinoma de Células Pequenas/metabolismo , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Feminino , Antígenos HLA/genética , Antígenos HLA/imunologia , Humanos , Imunoglobulinas/genética , Imunoglobulinas/imunologia , Interferon gama/imunologia , Interferon gama/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Proteínas de Membrana Transportadoras , Pessoa de Meia-Idade , Baço/citologia , Baço/imunologia , Baço/metabolismo , Microglobulina beta-2/genética , Microglobulina beta-2/imunologia
7.
Respir Res ; 7: 78, 2006 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-16709253

RESUMO

BACKGROUND: Effects of respiratory viral infection on airway epithelium include airway hyper-responsiveness and inflammation. Both features may contribute to the development of asthma. Excessive damage and loss of epithelial cells are characteristic in asthma and may result from viral infection. OBJECTIVE: To investigate apoptosis in Adenoviral-infected Guinea pigs and determine the role of death receptor and ligand expression in the airway epithelial response to limit viral infection. METHODS: Animal models included both an Acute and a Chronic Adeno-infection with ovalbumin-induced airway inflammation with/without corticosteroid treatment. Isolated airway epithelial cells were cultured to study viral production after infection under similar conditions. Immunohistochemistry, western blots and viral DNA detection were used to assess apoptosis, death receptor and TRAIL expression and viral release. RESULTS: In vivo and in vitro Adeno-infection demonstrated different apoptotic and death receptors (DR) 4 and 5 expression in response to corticosteroid exposure. In the Acute Adeno-infection model, apoptosis and DR4/5 expression was coordinated and were time-dependent. However, in vitro Acute viral infection in the presence of corticosteroids demonstrated delayed apoptosis and prolonged viral particle production. This reduction in apoptosis in Adeno-infected epithelial cells by corticosteroids exposure induced a prolonged virus production via both DR4 and TRAIL protein suppression. In the Chronic model where animals were ovalbumin-sensitized/challenged and were treated with corticosteroids, apoptosis was reduced relative to adenovirus-infected or corticosteroid alone. CONCLUSION: Our data suggests that apoptosis of infected cells limits viral production and may be mediated by DR4/5 and TRAIL expression. In the Acute model of Adeno-infection, corticosteroid exposure may prolong viral particle production by altering this apoptotic response of the infected cells. This results from decreased DR4 and TRAIL expression. In the Chronic model treated with corticosteroids, a similar decreased apoptosis was observed. This data suggests that DR and TRAIL modulation by corticosteroids may be important in viral infection of airway epithelium. The prolonged virus release in the setting of corticosteroids may result from reduced apoptosis and suppressed DR4/TRAIL expression by the infected cells.


Assuntos
Infecções por Adenoviridae/fisiopatologia , Infecções por Adenoviridae/virologia , Adenoviridae/crescimento & desenvolvimento , Anti-Inflamatórios/farmacologia , Apoptose , Budesonida/farmacologia , Traqueia/virologia , Doença Aguda , Infecções por Adenoviridae/complicações , Infecções por Adenoviridae/metabolismo , Animais , Células Cultivadas , Doença Crônica , Células Epiteliais/virologia , Feminino , Cobaias , Ovalbumina , Pneumonia/induzido quimicamente , Pneumonia/complicações , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Receptores do Fator de Necrose Tumoral/metabolismo , Traqueia/patologia , Traqueia/fisiopatologia , Vírion/fisiologia
8.
Cytotechnology ; 58(3): 173-9, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19101815

RESUMO

The expansion of stem cell numbers while retaining their developmental properties is a bioprocess challenge. We compared the growth rates and embryoid body (EB) formation yields of R1 and EFC murine embryonic stem cells (mESC) cultured in two basal media (DMEM or DMEM:F12) with additions of 1.7-15% fetal bovine serum (FBS) or serum replacer (KOSR). Whereas the basal medium or KOSR dose did not have a significant effect on growth rate for either cell line, increasing doses of KOSR had a significant negative effect on the EB yield of EFC cells. Use of DMEM:F12 and increasing doses of FBS independently and significantly increased the growth rate for both cell lines. DMEM:F12 also significantly increased EB yields for both cell lines. The results show that use of DMEM:F12 and several-fold lower than conventional concentrations of KOSR can efficiently support maintenance of mESC and that KOSR should be dose as well as lot optimized.

9.
PLoS One ; 3(6): e2469, 2008 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-18575595

RESUMO

BACKGROUND: Therapeutic intervention in many neurological diseases is thwarted by the physical obstacle formed by the blood-brain barrier (BBB) that excludes most drugs from entering the brain from the blood. Thus, identifying efficacious modes of drug delivery to the brain remains a "holy grail" in molecular medicine and nanobiotechnology. Brain capillaries, that comprise the BBB, possess an endogenous receptor that ferries an iron-transport protein, termed p97 (melanotransferrin), across the BBB. Here, we explored the hypothesis that therapeutic drugs "piggybacked" as conjugates of p97 can be shuttled across the BBB for treatment of otherwise inoperable brain tumors. APPROACH: Human p97 was covalently linked with the chemotherapeutic agents paclitaxel (PTAX) or adriamycin (ADR) and following intravenous injection, measured their penetration into brain tissue and other organs using radiolabeled and fluorescent derivatives of the drugs. In order to establish efficacy of the conjugates, we used nude mouse models to assess p97-drug conjugate activity towards glioma and mammary tumors growing subcutaneously compared to those growing intracranially. PRINCIPAL FINDINGS: Bolus-injected p97-drug conjugates and unconjugated p97 traversed brain capillary endothelium within a few minutes and accumulated to 1-2% of the injected by 24 hours. Brain delivery with p97-drug conjugates was quantitatively 10 fold higher than with free drug controls. Furthermore, both free-ADR and p97-ADR conjugates equally inhibited the subcutaneous growth of gliomas growing outside the brain. Evocatively, only p97-ADR conjugates significantly prolonged the survival of animals bearing intracranial gliomas or mammary tumors when compared to similar cumulated doses of free-ADR. SIGNIFICANCE: This study provides the initial proof of concept for p97 as a carrier capable of shuttling therapeutic levels of drugs from the blood to the brain for the treatment of neurological disorders, including classes of resident and metastatic brain tumors. It may be prudent, therefore, to consider implementation of this novel delivery platform in various clinical settings for therapeutic intervention in acute and chronic neurological diseases.


Assuntos
Antineoplásicos/administração & dosagem , Barreira Hematoencefálica , Portadores de Fármacos , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacocinética , Doxorrubicina/uso terapêutico , Corantes Fluorescentes , Humanos , Camundongos , Camundongos Nus , Paclitaxel/administração & dosagem , Paclitaxel/farmacocinética , Paclitaxel/uso terapêutico
10.
Int J Cancer ; 120(9): 1935-41, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17278102

RESUMO

Many immune therapeutic strategies are under development for melanoma to treat metastatic disease and prevent disease reoccurrence. However, human melanoma cells are often deficient in antigen processing and this appears to play a role in their expansion and escape from immunosurveillance. For example, expression of the transporters associated with antigen processing (TAP1 and TAP2) is down-regulated in the mouse melanoma cell line B16F10. This results in a lack of tumor-associated antigen processing, low surface expression of MHC Class I molecules and low immunogenicity. We observe that restoration of TAP1 expression by transfection resurrects the processing and presentation of viral antigens, and the melanoma-associated antigen, TRP-2. Immunization with irradiated B16F10/rTAP1 transfected cells generates CTLs that are capable of killing B16F10/rTAP1 transfected targets and B16F10 targets deficient in TAP1. Furthermore, B16F10/rTAP1 transfectants grow at a significantly slower rate in mice than B16F10 cells. In an experimental model that closely recapitulates the clinical situation, treatment of B16F10 tumors in mice with a vaccinia virus vector expressing TAP1 also significantly decreases tumor growth in vivo. Furthermore, tumors treated with vaccinia TAP1 had significantly reduced numbers of immunosuppressive, CD3(+)/IL-10 positive, tumor infiltrating lymphocytes. Therefore, TAP1 expression restores both antigen presentation and immunogenicity in B16F10 melanoma cells and concomitantly reduces immunosuppressive IL-10 production at the local tumor site, thereby increasing immunosurveillance mechanisms against tumors.


Assuntos
Transportadores de Cassetes de Ligação de ATP/fisiologia , Interleucina-10/biossíntese , Linfócitos do Interstício Tumoral/imunologia , Melanoma Experimental/imunologia , Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Apresentação de Antígeno , Antígenos H-2/análise , Antígeno de Histocompatibilidade H-2D , Antígenos de Histocompatibilidade Classe II/análise , Camundongos , Camundongos Endogâmicos C57BL
11.
Vaccine ; 25(12): 2331-9, 2007 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-17229499

RESUMO

Despite continued progress in understanding the pathophysiology of tumours, curative therapeutic options are still lacking for the metastatic form of the disease. One approach that has gathered considerable interest is the creation of therapeutic vaccines using genetically engineered non-replicating viruses as vehicles to revive immunosurveillance mechanisms that may eradicate residual tumour cells. A perceived problem with this approach is that the number of non-replicating viruses used as a vaccine inoculum does not remotely approximate the total number of cells in the body, nor even the number of tumour cells in the case of large tumour burden or metastasis. Here, we addressed the hypothesis that a limited amount of inoculum (1x10(8) PFU) of recombinant non-replicating adenovirus encoding human TAP1 (AdhTAP1) can induce protective immunity against 1.5x10(5) TAP-deficient, metastatic melanoma cells transplanted into a normal mouse (total of approximately 1x10(11) body cells). We show that efficacious anti-tumour cytolytic T cell responses are indeed induced by injecting melanoma-bearing animals with small numbers of recombinant viruses, resulting in increases in tumour-infiltrating dendritic cells, enhanced memory T cell subpopulations and, most importantly, in increased animal survival. This novel approach uses a limited input inoculum relative to the tumour cell mass, and thus achieves an efficacious outcome that has so far eluded other vaccine, immunotherapeutic or gene therapeutic strategies where there is a requisite for the majority of tumour cells to be transduced for beneficial outcome to be achieved.


Assuntos
Transportadores de Cassetes de Ligação de ATP/imunologia , Adenoviridae/genética , Vacinas Anticâncer/imunologia , Melanoma Experimental/imunologia , Linfócitos T/imunologia , Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/genética , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Feminino , Antígenos H-2/imunologia , Antígenos H-2/metabolismo , Humanos , Immunoblotting , Imunoterapia/métodos , Linfócitos do Interstício Tumoral/imunologia , Melanoma Experimental/patologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL
12.
J Exp Biol ; 208(Pt 1): 105-12, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15601882

RESUMO

Toads experiencing dehydrating conditions exhibit complex physiological and behavioral responses, some of which can potentially impact cutaneous gas exchange, an important component of total gas exchange. We measured the effect of dehydration on cutaneous gas exchange in the xeric-adapted toad Bufo woodhousei. First, two pharmacological agents were used to stimulate cutaneous blood flow--phentolamine (an alpha-blocker) and isoproterenol, a beta-stimulant and powerful cardio-accelerator--to determine a relationship between cutaneous blood flow and water loss. Both drugs increased heart rate and blood pressure, and caused visually evident extensive vasodilation of the skin. Untreated toads in a dry air stream took an average of 10.1+/-0.7 h to dehydrate to 80% body mass, while animals treated with isoproterenol and phentolamine requires only 7.2+/-0.8 h and 7.4+/-0.9 h, respectively. Rehydration, which was more rapid than dehydration, was similarly accelerated in pharmacologically treated toads. Cutaneous gas exchange (M(O2), M(CO2)) and C18O diffusing capacity (D(Skin)C18O) were then examined in unanesthetized toads under different states of body hydration. Blood gases and hematocrit were measured separately but under identical conditions. In fully hydrated toads at 23-25 degrees C, cutaneous gas exchange values were: M(O2) = 1.43+/-0.47 micromol g(-1) h(-1), M(CO2) = 1.75+/-0.85 micromol g(-1) h(-1), and the respiratory exchange ratio R = 1.36+/-0.56 (N=6, mean + 1 S.D.). D(Skin)C18O was 0.48+/-0.03 micromol g body mass(-1) h(-1) kPa. Following an enforced 20-25% loss of body water, D(Skin)C18O fell by nearly 50% to 0.28+/-0.09 micromol g(-1) h(-1) kPa. However, cutaneous M(O2), M(CO2) and R were unchanged at 1.48+/-0.15 micromol g(-1) h(-1), 1.72+/-0.29 micromol g(-1) h(-1) and 1.13+/-0.08 micromol g(-1) h(-1), respectively. Partial pressure of arterial (sciatic) oxygen, Pa(O2), normally about 12-13 kPa, remained unchanged by dehydration, but Pa(CO2) increased about 250% from 0.93+/-0.27 up to 2.27+/-0.93 kPa. The fall in D(Skin)C18O during dehydration presumably results at least in part from decreased cutaneous blood flow, possibly in an attempt to reduce the transcutaneous water loss that would otherwise result during dehydrating conditions. Concurrently, cutaneous M(CO2) is maintained under dehyrdating conditions by a greatly increased Pa(CO2) diffusion gradient across the skin. Thus, Bufo woodhousei appears able to restrict cutaneous blood flow without compromising vital cutaneous CO2 loss.


Assuntos
Adaptação Fisiológica , Bufonidae/fisiologia , Desidratação/fisiopatologia , Transporte Respiratório/fisiologia , Pele/irrigação sanguínea , Vasodilatação/efeitos dos fármacos , Perda Insensível de Água/fisiologia , Análise de Variância , Animais , Pressão Sanguínea/efeitos dos fármacos , Dióxido de Carbono/metabolismo , Isótopos de Carbono , Monóxido de Carbono/metabolismo , Frequência Cardíaca/efeitos dos fármacos , Hematócrito , Isoproterenol/farmacologia , Nevada , Fentolamina/farmacologia , Fluxo Sanguíneo Regional/fisiologia
13.
PLoS Pathog ; 1(4): e36, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16389301

RESUMO

We hypothesize that over-expression of transporters associated with antigen processing (TAP1 and TAP2), components of the major histocompatibility complex (MHC) class I antigen-processing pathway, enhances antigen-specific cytotoxic activity in response to viral infection. An expression system using recombinant vaccinia virus (VV) was used to over-express human TAP1 and TAP2 (VV-hTAP1,2) in normal mice. Mice coinfected with either vesicular stomatitis virus plus VV-hTAP1,2 or Sendai virus plus VV-hTAP1,2 increased cytotoxic lymphocyte (CTL) activity by at least 4-fold when compared to coinfections with a control vector, VV encoding the plasmid PJS-5. Coinfections with VV-hTAP1,2 increased virus-specific CTL precursors compared to control infections without VV-hTAP1,2. In an animal model of lethal viral challenge after vaccination, VV-hTAP1,2 provided protection against a lethal challenge of VV at doses 100-fold lower than control vector alone. Mechanistically, the total MHC class I antigen surface expression and the cross-presentation mechanism in spleen-derived dendritic cells was augmented by over-expression of TAP. Furthermore, VV-hTAP1,2 increases splenic TAP transport activity and endogenous antigen processing, thus rendering infected targets more susceptible to CTL recognition and subsequent killing. This is the first demonstration that over-expression of a component of the antigen-processing machinery increases endogenous antigen presentation and dendritic cell cross-presentation of exogenous antigens and may provide a novel and general approach for increasing immune responses against pathogens at low doses of vaccine inocula.

14.
J Virol ; 76(4): 1578-87, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11799152

RESUMO

E3-6.7K is a small and hydrophobic membrane glycoprotein encoded by the E3 region of subgroup C adenovirus. Recently, E3-6.7K has been shown to be required for the downregulation of tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) receptors by the adenovirus E3/10.4K and E3/14.5K complex of proteins. We demonstrate here that E3-6.7K has additional protective roles, independent of other virus proteins. In transfected Jurkat T-cell lymphoma cells, E3-6.7K was found to maintain endoplasmic reticulum-Ca(2+) homeostasis and inhibit the induction of apoptosis by thapsigargin. The presence of E3-6.7K also lead to a reduction in the TNF-induced release of arachidonic acid from transfected U937 human histiocytic lymphoma cells. In addition, E3-6.7K protected cells against apoptosis induced through Fas, TNF receptor, and TRAIL receptors. Therefore, E3-6.7K confers a wide range of protective effects against both Ca(2+) flux-induced and death receptor-mediated apoptosis.


Assuntos
Proteínas E3 de Adenovirus/farmacologia , Apoptose/efeitos dos fármacos , Ácido Araquidônico/metabolismo , Cálcio/fisiologia , Homeostase/efeitos dos fármacos , Proteínas de Membrana , Proteínas E3 de Adenovirus/genética , Adenovírus Humanos/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Células Jurkat , Tapsigargina/farmacologia , Transfecção
15.
Am J Respir Cell Mol Biol ; 26(1): 52-7, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11751203

RESUMO

This study was designed to test the hypothesis that cigarette smoke-induced inflammation and emphysema are amplified by the presence of latent adenoviral (Ad) infection, and to determine whether this emphysematous process can be reversed by all-trans-retinoic acid (RA) treatment. The results confirm that in guinea pigs, chronic cigarette-smoke exposure caused lesions similar to human centrilobular emphysema. They also show that latent Ad infection combined with cigarette-smoke exposure caused an excess increase in lung volume (P < 0.001), air-space volume (P < 0.001), and lung weight (P < 0.01), and further decrease in surface-to-volume ratio (P < 0.001) compared with smoke exposure alone. RA treatment failed to reverse these emphysematous changes. Analysis of inflammatory response in parenchymal and airway tissue showed that smoking caused an increase of polymorphonuclear leukocytes (PMNs) (P < 0.0002), macrophages (P < 0.001), and CD4 cells (P < 0.0009), and that latent Ad infection independently increased PMNs (P < 0.001), macrophages (P = 0.003), and CD8 cells (P < 0.001). We conclude that latent Ad infection amplifies the emphysematous lung destruction and increases the inflammatory response produced by cigarette-smoke exposure. In this study, the increase in CD4 was associated with cigarette smoke and the increase in CD8 cells with latent Ad infection.


Assuntos
Adenoviridae/genética , Enfisema/induzido quimicamente , Inflamação , Pulmão/patologia , Pulmão/virologia , Fumar , Análise de Variância , Animais , Peso Corporal , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Cromatografia Líquida de Alta Pressão , Feminino , Cobaias , Pulmão/imunologia , Macrófagos/metabolismo , Neutrófilos/metabolismo , Tamanho do Órgão , Tretinoína/sangue , Tretinoína/farmacologia
16.
Microcirculation ; 10(6): 457-62, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14745458

RESUMO

OBJECTIVE: This study was undertaken to assess the role of p97 (also known as melanotransferrin) in the transfer of iron into the brain, because the passage of most large molecules is limited by the presence of the blood-brain barrier, including that of the serum iron transporter transferrin. METHODS: To study the function of the soluble form of p97, we followed the uptake of radioiodinated and 55Fe loaded p97 and transferrin by the brain during a 24-hour period. RESULTS: We show that the soluble form of p97 has the ability to transcytose across the murine blood-brain barrier, and its transcytosis can be inhibited in a specific manner. We also provide evidence that p97 transports iron into the brain more efficiently than transferrin. CONCLUSIONS: These data support the idea that p97 is an important iron transporter across the blood-brain barrier in normal physiology and possibly in neurodegenerative diseases, such as Alzheimer disease, in which iron homeostasis in the brain becomes disrupted.


Assuntos
Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Ferro/metabolismo , Proteínas de Neoplasias/farmacocinética , Animais , Antígenos de Neoplasias , Transporte Biológico , Humanos , Radioisótopos do Iodo , Radioisótopos de Ferro , Antígenos Específicos de Melanoma , Camundongos , Camundongos Endogâmicos C57BL , Transferrina/farmacocinética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa