Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-38586014

RESUMO

Current COVID-19 mRNA vaccines delivered intramuscularly (IM) induce effective systemic immunity, but with suboptimal immunity at mucosal sites, limiting their ability to impart sterilizing immunity. There is strong interest in rerouting immune responses induced in the periphery by parenteral vaccination to the portal entry site of respiratory viruses, such as SARS-CoV-2, by mucosal vaccination. We previously demonstrated the combination adjuvant, NE/IVT, consisting of a nanoemulsion (NE) and an RNA-based RIG-I agonist (IVT) induces potent systemic and mucosal immune responses in protein-based SARS-CoV-2 vaccines administered intranasally (IN). Herein, we demonstrate priming IM with mRNA followed by heterologous IN boosting with NE/IVT adjuvanted recombinant antigen induces strong mucosal and systemic antibody responses and enhances antigen-specific T cell responses in mucosa-draining lymph nodes compared to IM/IM and IN/IN prime/boost regimens. While all regimens induced cross-neutralizing antibodies against divergent variants and sterilizing immunity in the lungs of challenged mice, mucosal vaccination, either as homologous prime/boost or heterologous IN boost after IM mRNA prime was required to impart sterilizing immunity in the upper respiratory tract. Our data demonstrate the benefit of hybrid regimens whereby strong immune responses primed via IM vaccination are rerouted by IN vaccination to mucosal sites to provide optimal protection to SARS-CoV-2.

2.
Cancer Gene Ther ; 31(3): 464-471, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38177307

RESUMO

KRAS mutations occur commonly in the lung and can lead to the development of non-small cell lung cancer (NSCLC). While the mutated KRAS protein is a neoantigen, it usually does not generate an effective anti-tumor immune response on mucosal/epithelial surfaces. Despite this, mutated KRAS remains a potential target for immunotherapy since immune targeting of this protein in animal models has been effective at eliminating tumor cells. We attempted to develop a KRAS vaccine using mutated and wild-type KRAS peptides in combination with a nanoemulsion (NE) adjuvant. The efficacy of this approach was tested in an inducible mutant KRAS-mouse lung tumor model. Animals were immunized intranasally using NE with KRAS peptides. These animals had decreased CD4+FoxP3+ T cells in both lymph nodes and spleen. Immunized animals also showed higher IFN-γ and IL-17a levels to mutated KRAS that were produced by CD8+ T cells and enhancement in KRAS-specific Th1 and Th17 responses that persisted for 3 months after the last vaccination. Importantly, the immunized animals had significantly decreased tumor incidence compared to control animals. In conclusion, a mucosal approach to KRAS vaccination demonstrated the ability to induce local KRAS-specific immune responses in the lung and resulted in reduced tumor incidence.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Camundongos , Animais , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/terapia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/prevenção & controle , Vacinas de Subunidades Proteicas , Proteínas Proto-Oncogênicas p21(ras)/genética , Linfócitos T CD8-Positivos , Modelos Animais de Doenças , Peptídeos/genética , Mutação
3.
Pharmaceutics ; 15(7)2023 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-37514184

RESUMO

The objective of the study was to explore the feasibility of a new drug delivery system using laponite (LAP) and cyclic poly(ethylene glycol) (cPEG). Variously shaped and flexible hybrid nanocrystals were made by both the covalent and physical attachment of chemically homogeneous cyclized PEG to laponite nanodisc plates. The size of the resulting, nearly spherical particles ranged from 1 to 1.5 µm, while PEGylation with linear methoxy poly (ethylene glycol) (mPEG) resulted in fragile sheets of different shapes and sizes. When infused with 10% doxorubicin (DOX), a drug commonly used in the treatment of various cancers, the LAP-cPEG/DOX formulation was transparent and maintained liquid-like homogeneity without delamination, and the drug loading efficiency of the LAP-cPEG nano system was found to be higher than that of the laponite-poly(ethylene glycol) LAP-mPEG system. Furthermore, the LAP-cPEG/DOX formulation showed relative stability in phosphate-buffered saline (PBS) with only 15% of the drug released. However, in the presence of human plasma, about 90% of the drug was released continuously over a period of 24 h for the LAP-cPEG/DOX, while the LAP-mPEG/DOX formulation released 90% of DOX in a 6 h burst. The results of the cell viability assay indicated that the LAP-cPEG/DOX formulation could effectively inhibit the proliferation of A549 lung carcinoma epithelial cells. With the DOX concentration in the range of 1-2 µM in the LAP-cPEG/DOX formulation, enhanced drug effects in both A549 lung carcinoma epithelial cells and primary lung epithelial cells were observed compared to LAP-mPEG/DOX. The unique properties and effects of cPEG nanoparticles provide a potentially better drug delivery system and generate interest for further targeting studies and applications.

4.
Immunohorizons ; 7(6): 480-492, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37341756

RESUMO

Vitamin A and its biologically active metabolites, all-trans and 9-cis retinoic acid (RA), are thought to be important in generating and modulating immune function. However, RA modulates the function of many types of immune cells, and its specific role in dendritic cell (DC) activation, Ag presentation, and T cell effector function has not been fully characterized. Because RA works primarily through RA receptor (RAR)α, we examined mice with a myeloid cell-specific defect in RA signaling. These transgenic mice have a CD11c-cre-driven expression of a truncated form of RARα that specifically blocks the signaling of all forms of RARs in myeloid cells. This defect results in abnormal DC function, with impaired DC maturation and activation, and reduced Ag uptake and processing. These DC abnormalities were associated with a reduced ability to mount Ag-specific T cell responses to immunization despite having normally functioning T cells. In contrast, the loss of DC-specific RA signaling did not significantly alter levels of Ag-specific Abs postimmunization and resulted in an increase in bronchial IgA. Our findings indicate that RA signaling in DCs is crucial for immune activation, and its absence impairs the development of Ag-specific effector functions of T cell immunity.


Assuntos
Linfócitos T , Tretinoína , Camundongos , Animais , Tretinoína/farmacologia , Tretinoína/metabolismo , Linfócitos T/metabolismo , Diferenciação Celular , Transdução de Sinais , Camundongos Transgênicos , Células Dendríticas
5.
Molecules ; 28(5)2023 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-36903437

RESUMO

Therapeutics, based on small interfering RNA (siRNA), have demonstrated tremendous potential for treating cancer. However, issues such as non-specific targeting, premature degradation, and the intrinsic toxicity of the siRNA, have to be solved before they are ready for use in translational medicines. To address these challenges, nanotechnology-based tools might help to shield siRNA and ensure its specific delivery to the target site. Besides playing a crucial role in prostaglandin synthesis, the cyclo-oxygenase-2 (COX-2) enzyme has been reported to mediate carcinogenesis in various types of cancer, including hepatocellular carcinoma (HCC). We encapsulated COX-2-specific siRNA in Bacillus subtilis membrane lipid-based liposomes (subtilosomes) and evaluated their potential in the treatment of diethylnitrosamine (DEN)-induced hepatocellular carcinoma. Our findings suggested that the subtilosome-based formulation was stable, releasing COX-2 siRNA in a sustained manner, and has the potential to abruptly release encapsulated material at acidic pH. The fusogenic property of subtilosomes was revealed by FRET, fluorescence dequenching, content-mixing assay, etc. The subtilosome-based siRNA formulation was successful in inhibiting TNF-α expression in the experimental animals. The apoptosis study indicated that the subtilosomized siRNA inhibits DEN-induced carcinogenesis more effectively than free siRNA. The as-developed formulation also suppressed COX-2 expression, which in turn up-regulated the expression of wild-type p53 and Bax on one hand and down-regulated Bcl-2 expression on the other. The survival data established the increased efficacy of subtilosome-encapsulated COX-2 siRNA against hepatocellular carcinoma.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animais , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/patologia , Dietilnitrosamina/farmacologia , RNA Interferente Pequeno/farmacologia , Ciclo-Oxigenase 2 , Apoptose , Carcinogênese
6.
J Allergy Clin Immunol ; 150(1): 1-11, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35569567

RESUMO

Recent events involving the global coronavirus pandemic have focused attention on vaccination strategies. Although tremendous advances have been made in subcutaneous and intramuscular vaccines during this time, one area that has lagged in implementation is mucosal immunization. Mucosal immunization provides several potential advantages over subcutaneous and intramuscular routes, including protection from localized infection at the site of entry, clearance of organisms on mucosal surfaces, induction of long-term immunity through establishment of central and tissue-resident memory cells, and the ability to shape regulatory responses. Despite these advantages, significant barriers remain to achieving effective mucosal immunization. The epithelium itself provides many obstacles to immunization, and the activation of immune recognition and effector pathways that leads to mucosal immunity has been difficult to achieve. This review will highlight the potential advantages of mucosal immunity, define the barriers to mucosal immunization, examine the immune mechanisms that need to be activated on mucosal surfaces, and finally address recent developments in methods for mucosal vaccination that have shown promise in generating immunity on mucosal surfaces in human trials.


Assuntos
Imunização , Vacinas , Humanos , Imunidade nas Mucosas , Imunização/métodos , Mucosa , Vacinação/métodos
7.
Front Immunol ; 12: 599296, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33717078

RESUMO

We have demonstrated that intranasal immunotherapy with allergens formulated in a nanoemulsion (NE) mucosal adjuvant suppresses Th2/IgE-mediated allergic responses and protects from allergen challenge in murine food allergy models. Protection conferred by this therapy is associated with strong suppression of allergen specific Th2 cellular immunity and increased Th1 cytokines. Here we extend these studies to examine the effect of NE-allergen immunization in mice sensitized to multiple foods. Mice were sensitized to both egg and peanut and then received NE vaccine formulated with either one or both of these allergens. The animals were then subjected to oral challenges with either egg or peanut to assess reactivity. Immunization with NE formulations containing both egg and peanut markedly reduced reactivity after oral allergen challenge with either allergen. Interestingly, mice that received the vaccine containing only peanut also had reduced reactivity to challenge with egg. Protection from oral allergen challenge was achieved despite the persistence of allergen-specific IgE and was associated with strong suppression of both Th2-polarized immune responses, alarmins and type 2 innate lymphoid cells (ILC2). NE-induced bystander suppression of reactivity required IFN-γ and the presence of an allergen in the NE vaccine. These results demonstrate that anaphylactic reactions to food allergens can be suppressed using allergen-specific immunotherapy without having to eliminate allergen-specific IgE and suggests that modulation of Th2 immunity towards one allergen may induce bystander effects that suppress reactivity to other allergens through the induction of IFN-γ and suppression of alarmins in the intestine. In addition, these data suggest that a NE vaccine for a single food allergen may lead to a global suppression of allergic responses to multiple foods.


Assuntos
Alarminas/genética , Alérgenos/imunologia , Hipersensibilidade Alimentar/etiologia , Hipersensibilidade Alimentar/terapia , Regulação da Expressão Gênica , Vacinas/imunologia , Adjuvantes Imunológicos , Administração Intranasal , Animais , Efeito Espectador , Citocinas/metabolismo , Dessensibilização Imunológica , Modelos Animais de Doenças , Imunoglobulina E/imunologia , Imunomodulação , Camundongos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Vacinas/administração & dosagem
8.
Front Immunol ; 10: 916, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31134057

RESUMO

Mucosal surfaces are the primary point of entry for many infectious agents and mucosal immune responses serve as the primary defense to these pathogens. In order to mount an effective mucosal immune response, it is important to induce T cell homing to mucosal surfaces. Conventional vaccine adjuvants induce strong systemic immunity but often fail to produce mucosal immunity. We have developed an oil-in-water nanoemulsion (NE) adjuvant that provides mucosal immunity and efficient protection against mucosal pathogens when administered as part of an intranasal vaccine. In the present study, we demonstrate that intranasal immunization with NE indirectly activates the retinaldehyde dehydrogenase (RALDH) activity in dendritic cells through epithelial cell activity leading to SIgA as well as potent cellular responses and expression of α4ß7 and CCR9 gut homing receptors on T cells. Confirming these findings, ex-vivo stimulation of splenocytes from NE nasally immunized animals showed increase in Th1/Th17 cytokines while suppressing Th2 responses. In examining mechanisms underlying this activation NE activated RALDH via MyD88 dependent pathways in DCs but did not activate the retinoic acid receptor directly. These results suggest that RALDH immune activities can be achieved by epithelial activation without direct RAR activation, which has significant implications for understanding mucosal immunity and the design of mucosal vaccines.


Assuntos
Adjuvantes Imunológicos/farmacologia , Células Dendríticas/imunologia , Imunidade nas Mucosas/efeitos dos fármacos , Fator 88 de Diferenciação Mieloide/imunologia , Nanoestruturas , Retinal Desidrogenase/imunologia , Transdução de Sinais/efeitos dos fármacos , Administração Intranasal , Animais , Linhagem Celular , Emulsões , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/imunologia , Camundongos , Transdução de Sinais/imunologia
9.
PLoS One ; 13(7): e0199612, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29975735

RESUMO

Acute exacerbations are the major cause of morbidity and mortality in patients with chronic obstructive pulmonary disease (COPD). Rhinovirus, which causes acute exacerbations may also accelerate progression of lung disease in these patients. Current therapies reduces the respiratory symptoms and does not treat the root cause of exacerbations effectively. We hypothesized that quercetin, a potent antioxidant and anti-inflammatory agent with antiviral properties may be useful in treating rhinovirus-induced changes in COPD. Mice with COPD phenotype maintained on control or quercetin diet and normal mice were infected with sham or rhinovirus, and after 14 days mice were examined for changes in lung mechanics and lung inflammation. Rhinovirus-infected normal mice showed no changes in lung mechanics or histology. In contrast, rhinovirus-infected mice with COPD phenotype showed reduction in elastic recoiling and increase in lung inflammation, goblet cell metaplasia, and airways cholinergic responsiveness compared to sham-infected mice. Interestingly, rhinovirus-infected mice with COPD phenotype also showed accumulation of neutrophils, CD11b+/CD11c+ macrophages and CD8+ T cells in the lungs. Quercetin supplementation attenuated rhinovirus-induced all the pathologic changes in mice with COPD phenotype. Together these results indicate that quercetin effectively mitigates rhinovirus-induced progression of lung disease in a mouse model of COPD. Therefore, quercetin may be beneficial in the treatment of rhinovirus-associated exacerbations and preventing progression of lung disease in COPD.


Assuntos
Infecções por Picornaviridae/complicações , Infecções por Picornaviridae/virologia , Doença Pulmonar Obstrutiva Crônica/etiologia , Doença Pulmonar Obstrutiva Crônica/patologia , Quercetina/farmacologia , Rhinovirus , Animais , Anti-Inflamatórios/farmacologia , Antivirais/farmacologia , Biópsia , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Mediadores da Inflamação/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Infiltração de Neutrófilos , Neutrófilos/metabolismo , Neutrófilos/patologia , Fenótipo , Infecções por Picornaviridae/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/fisiopatologia , Rhinovirus/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/patologia
10.
J Immunol ; 197(6): 2409-20, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27503209

RESUMO

Airway epithelial cells are the major target for rhinovirus (RV) infection and express proinflammatory chemokines and antiviral cytokines that play a role in innate immunity. Previously, we demonstrated that RV interaction with TLR2 causes ILR-associated kinase-1 (IRAK-1) depletion in both airway epithelial cells and macrophages. Further, IRAK-1 degradation caused by TLR2 activation was shown to inhibit ssRNA-induced IFN expression in dendritic cells. Therefore, in this study, we examined the role of TLR2 and IRAK-1 in RV-induced IFN-ß, IFN-λ1, and CXCL-10, which require signaling by viral RNA. In airway epithelial cells, blocking TLR2 enhanced RV-induced expression of IFNs and CXCL-10. By contrast, IRAK-1 inhibition abrogated RV-induced expression of CXCL-10, but not IFNs in these cells. Neutralization of IL-33 or its receptor, ST2, which requires IRAK-1 for signaling, inhibited RV-stimulated CXCL-10 expression. In addition, RV induced expression of both ST2 and IL-33 in airway epithelial cells. In macrophages, however, RV-stimulated CXCL-10 expression was primarily dependent on TLR2/IL-1R. Interestingly, in a mouse model of RV infection, blocking ST2 not only attenuated RV-induced CXCL-10, but also lung inflammation. Finally, influenza- and respiratory syncytial virus-induced CXCL-10 was also found to be partially dependent on IL-33/ST2/IRAK-1 signaling in airway epithelial cells. Together, our results indicate that RV stimulates CXCL-10 expression via the IL-33/ST2 signaling axis, and that TLR2 signaling limits RV-induced CXCL-10 via IRAK-1 depletion at least in airway epithelial cells. To our knowledge, this is the first report to demonstrate the role of respiratory virus-induced IL-33 in the induction of CXCL-10 in airway epithelial cells.


Assuntos
Quimiocina CXCL10/imunologia , Células Epiteliais/imunologia , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Mucosa Respiratória/imunologia , Rhinovirus/imunologia , Receptor 2 Toll-Like/imunologia , Animais , Brônquios/citologia , Brônquios/imunologia , Células Cultivadas , Quimiocina CXCL10/genética , Quimiocinas/imunologia , Citocinas/imunologia , Células Epiteliais/virologia , Humanos , Imunidade Inata , Proteína 1 Semelhante a Receptor de Interleucina-1/imunologia , Interleucina-33/imunologia , Camundongos , Infecções por Picornaviridae/imunologia , Infecções por Picornaviridae/virologia , Mucosa Respiratória/virologia , Transdução de Sinais , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo
11.
Arterioscler Thromb Vasc Biol ; 36(2): 380-8, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26743170

RESUMO

OBJECTIVE: Increased vascular permeability is a hallmark of sepsis and acute respiratory distress syndrome. Angiopoietin (Ang2) induces vascular leak, and excess Ang2 generation is associated with patient mortality from these diseases. However, mechanisms dampening Ang2 generation during injury remain unclear. Interestingly, microRNA (miR)-150 levels were decreased in septic patients. miR regulate signaling networks by silencing mRNAs containing complementary sequences. Thus, we hypothesized that miR-150 suppresses Ang2 generation and thereby resolves vascular injury. APPROACH AND RESULTS: Wild-type or miR-150(-/-) mice or endothelial cells were exposed to lipopolysaccharide or sepsis, and Ang2 levels, adherens junction reannealing, endothelial barrier function, and mortality were determined. Although Ang2 transiently increased during lipopolysaccharide-induced injury in wild-type endothelial cells and lungs, miR-150 expression was elevated only during recovery from injury. Deletion of miR-150 caused a persistent increase in Ang2 levels and impaired adherens junctions reannealing after injury, resulting thereby in an irreversible increase in vascular permeability. Also, miR-150(-/-) mice died rapidly after sepsis. Rescuing miR-150 expression in endothelial cells prevented Ang2 generation, thereby restoring vascular barrier function in miR-150(-/-) mice. miR-150 terminated Ang2 generation by targeting the transcription factor, early growth response 2. Thus, early growth response 2 or Ang2 depletion in miR-150(-/-) endothelial cells restored junctional reannealing and reinstated barrier function. Importantly, upregulating miR-150 expression by injecting a chemically synthesized miR-150 mimic into wild-type mice vasculature decreased early growth response 2 and Ang2 levels and hence mortality from sepsis. CONCLUSIONS: miR-150 is a novel suppressor of Ang2 generation with a key role in resolving vascular injury and reducing mortality resulting from sepsis.


Assuntos
Angiopoietina-2/metabolismo , Células Endoteliais/metabolismo , MicroRNAs/metabolismo , Artéria Pulmonar/metabolismo , Doenças Vasculares/metabolismo , Remodelação Vascular , Junções Aderentes/metabolismo , Junções Aderentes/patologia , Angiopoietina-2/genética , Animais , Permeabilidade Capilar , Células Cultivadas , Modelos Animais de Doenças , Proteína 2 de Resposta de Crescimento Precoce/metabolismo , Células Endoteliais/patologia , Regulação da Expressão Gênica , Genótipo , Lipopolissacarídeos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , Fenótipo , Artéria Pulmonar/patologia , Sepse/complicações , Transdução de Sinais , Fatores de Tempo , Transfecção , Doenças Vasculares/etiologia , Doenças Vasculares/genética , Doenças Vasculares/patologia , Doenças Vasculares/terapia
12.
FASEB J ; 30(1): 102-10, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26316271

RESUMO

Stability of endothelial cell (EC) adherens junctions (AJs) is central for prevention of tissue edema, the hallmark of chronic inflammatory diseases including acute respiratory distress syndrome. Here, we demonstrate a previously unsuspected role of sphingosine kinase 1 (SPHK1) in the mechanism by which transient receptor potential channel 1 (Trpc1)-mediated Ca(2+) entry destabilizes AJs. Trpc1(-/-) monolayers showed a 2.2-fold increase in vascular endothelial (VE)-cadherin cell-surface expression above wild-type (WT) monolayers. Thrombin increased endothelial permeability (evident by a 5-fold increase in interendothelial gap area and 60% decrease in transendothelial electrical resistance) in WT but not Trpc1(-/-) ECs. Trpc1(-/-) mice resisted the hyperpermeability effects of the edemagenic agonists used and exhibited 60% less endotoxin-induced mortality. Because sphingosine-1-phosphate (S1P) strengthens AJs, we determined if TRPC1 functioned by inhibiting SPHK1 activity, which generates S1P. Intriguingly, Trpc1(-/-) ECs or ECs transducing a TRPC1-inactive mutant showed a 1.5-fold increase in basal SPHK1 expression compared with WT ECs, resulting in a 2-fold higher S1P level. SPHK1 inhibitor SK1-I decreased basal transendothelial electrical resistance more in WT ECs (48 and 72% reduction at 20 and 50 µM, respectively) than in Trpc1(-/-) ECs. However, SK1-I pretreatment rescued thrombin-induced EC permeability in Trpc1(-/-) ECs. Thus, TRPC1 suppression of basal SPHK1 activity enables EC-barrier destabilization by edemagenic agonists.


Assuntos
Junções Aderentes/metabolismo , Células Endoteliais/metabolismo , Endotélio/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Canais de Cátion TRPC/metabolismo , Animais , Caderinas/metabolismo , Cálcio/metabolismo , Permeabilidade da Membrana Celular , Camundongos Knockout , Transdução de Sinais/fisiologia , Canais de Cátion TRPC/genética
13.
Int J Nanomedicine ; 9: 1139-52, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24627632

RESUMO

Curcumin (diferuloylmethane) is found in large quantities in the roots of Curcuma longa. It possesses strong antioxidant and anti-inflammatory properties, and inhibits chemically-induced carcinogenesis in the skin, forestomach, colon, and liver. Unfortunately, the poor bioavailability and hydrophobicity of curcumin pose a major hurdle to its use as a potent anticancer agent. To circumvent some of these problems, we developed a novel, dual-core microcell formulation of curcumin. The encapsulation of curcumin in microcells increases its solubility and bioavailability, and facilitates slow release kinetics over extended periods. Besides being safe, these formulations do not bear any toxicity constraints, as revealed by in vitro and in vivo studies. Histopathological analysis revealed that curcumin-bearing microcells helped in regression of hepatocellular carcinoma and the maintenance of cellular architecture in liver tissue. Free curcumin had a very mild effect on cancer suppression. Empty (sham) microcells and microparticles failed to inhibit cancer cells. The novel curcumin formulation was found to suppress hepatocellular carcinoma efficiently in Swiss albino mice.


Assuntos
Antineoplásicos Fitogênicos/administração & dosagem , Curcumina/administração & dosagem , Sistemas de Liberação de Medicamentos , Neoplasias Hepáticas Experimentais/tratamento farmacológico , Fitoterapia , Animais , Antineoplásicos Fitogênicos/farmacocinética , Antineoplásicos Fitogênicos/toxicidade , Disponibilidade Biológica , Sobrevivência Celular/efeitos dos fármacos , Curcumina/farmacocinética , Curcumina/toxicidade , Composição de Medicamentos/métodos , Feminino , Células Hep G2 , Hepatócitos/efeitos dos fármacos , Humanos , Ácido Láctico/química , Neoplasias Hepáticas Experimentais/metabolismo , Neoplasias Hepáticas Experimentais/patologia , Camundongos , Nanomedicina , Nanopartículas/química , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Fator de Necrose Tumoral alfa/metabolismo
14.
Nanomedicine ; 9(2): 247-56, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22858760

RESUMO

In the current study, a novel niosome based formulation of diallyl disulfide (DADS) was evaluated for its potential to treat disseminated candidiasis in mouse model. Among various non-ionic surfactants tested, niosome formulation prepared using Span 80 was found to be most efficient in the entrapment of DADS. The DADS loaded niosomes had size dimensions in the range of 140 ± 30 nm with zeta potential of -30.67 ± 4.5. Liver/kidney function tests as well as histopathologic studies suggested that noisome-based DADS formulations are safe at the dose investigated. When administered to Candida albicans infected animals, the DADS bearing niosomal formulation cleared the fungal burden and increased their survival much efficiently than its free form. FROM THE CLINICAL EDITOR: In this study, a novel niosomal formulation of the antifungal DADS was utilized in a murine candidiasis model, resulting in more efficient fungal clearance compared to the free formulation.


Assuntos
Compostos Alílicos/uso terapêutico , Antifúngicos/uso terapêutico , Candida albicans/efeitos dos fármacos , Candidíase/tratamento farmacológico , Dissulfetos/uso terapêutico , Portadores de Fármacos/química , Hexoses/química , Tensoativos/química , Compostos Alílicos/administração & dosagem , Compostos Alílicos/farmacocinética , Animais , Antifúngicos/administração & dosagem , Antifúngicos/farmacocinética , Dissulfetos/administração & dosagem , Dissulfetos/farmacocinética , Portadores de Fármacos/toxicidade , Feminino , Hexoses/toxicidade , Camundongos , Camundongos Endogâmicos BALB C , Tensoativos/toxicidade
15.
Int J Nanomedicine ; 7: 35-47, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22275821

RESUMO

In the present study, a novel poly-lactic glycolic acid (PLGA)-based microparticle formulation of perillyl alcohol (POH) was prepared and characterized. Further, its efficacy was evaluated against di-methyl benzo anthracene-induced skin papilloma in Swiss albino mice. The characterization studies showed that POH-bearing PLGA microparticles were of the size 768 ± 215 nm with a ζ-potential value of -7.56 ± 0.88 mV. The entrapment efficiency of the active drug in particles was 42.4% ± 3.5%. POH-bearing PLGA microparticles were stable and released entrapped drug gradually over an extended time period. The in vitro efficacy of POH-bearing PLGA microparticles was evaluated by examining their differential cytotoxicity and assessing their ability to inhibit epidermoid carcinoma cell line (A253). The POH-based microparticles when administered to tumor-bearing animals caused greater tumor regression and increased survival rate (∼80%) as compared with the group receiving free form of POH (survival rate 40%). The superiority of POH-PLGA microparticles over free form of POH was further evident from their ability to modulate apoptosis-regulating factors.


Assuntos
Antineoplásicos/farmacologia , Ácido Láctico/farmacologia , Microesferas , Monoterpenos/farmacologia , Papiloma/tratamento farmacológico , Ácido Poliglicólico/farmacologia , Neoplasias Cutâneas/tratamento farmacológico , Análise de Variância , Animais , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose , Linhagem Celular Tumoral , Feminino , Estimativa de Kaplan-Meier , Ácido Láctico/química , Camundongos , Monoterpenos/química , Papiloma/induzido quimicamente , Papiloma/patologia , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Biosci Rep ; 31(4): 265-72, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20687896

RESUMO

The present study was aimed at evaluating the effectiveness of amoxicillin-bearing HSA (human serum albumin) and PLGA [poly(lactic-co-glycolic acid)] microparticles in combating Listeria monocytogenes infection in Swiss albino mice. Amoxicillin-bearing HSA microspheres were prepared by chemical cross-linking of a drug/albumin mixture with glutaraldehyde, and PLGA microspheres were prepared by the W/O/W (water-in-oil-in-water) emulsion technique. The microspheres were characterized for their size, ζ potential and entrapment efficiency using SEM (scanning electron microscopy) and a Zetasizer. Release kinetics was performed in a phosphate buffer (pH 7.4) at 37°C simulating physiological conditions. Bacterial burden in various vital organs and survival data established enhanced efficacy of PLGA and HSA microspheres as compared with free drug. Among the two delivery systems, PLGA microspheres, when compared with HSA microspheres, imparted better efficacy in terms of reduction in bacterial load as well as increase in survival. The results of the present study clearly demonstrate that microparticles successfully target the infected macrophages and the approach could be well exploited for targeting the intracellular pathogens as well.


Assuntos
Amoxicilina/administração & dosagem , Antibacterianos/administração & dosagem , Portadores de Fármacos/química , Ácido Láctico/química , Listeriose/tratamento farmacológico , Ácido Poliglicólico/química , Amoxicilina/química , Amoxicilina/uso terapêutico , Animais , Antibacterianos/química , Antibacterianos/uso terapêutico , Sistemas de Liberação de Medicamentos , Feminino , Listeria monocytogenes/efeitos dos fármacos , Camundongos , Microscopia Eletrônica de Varredura , Tamanho da Partícula , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Polímeros/química
17.
Biosci Rep ; 30(4): 223-31, 2010 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-19642969

RESUMO

In the present study, we evaluated the potential of a liposomal formulation of the garlic oil component DAS (diallyl sulfide) in treating disseminated infection caused by the intracellular opportunistic pathogen Candida albicans in experimental mice. The PC (phosphatidylcholine) liposomal formulation of DAS was evaluated for size, zeta-potential, entrapment efficiency and release kinetics, toxicity etc. For therapeutic studies, mice were challenged with intravenous infection dosage of 10(7) blastospores of C. albicans followed by treatment with various doses of DAS formulations [12 and 6 mg/kg b.w. (body mass)] three times, on alternative days. The antifungal efficacy of liposomal DAS was assessed on the basis of survival of treated mice as well as the residual fungal load in vital organs like liver and spleen of mice. The results of the present study showed that treatment with DAS-bearing liposomes (12 mg/kg b.w.) resulted in the highest survival rate in animals. Liposomal DAS also significantly decreased residual fungal load in vital organs of experimental animals compared with the free form of DAS. The liposomal DAS was also found to be free of toxic manifestations as revealed by the erythrocyte lysis test and liver/kidney function tests. The results of the present study established that the antifungal activity of DAS, a poorly soluble compound, can be enhanced by the incorporation of it into liposomes. Further studies and optimizations are needed to build upon the promising findings of this study to enable the development of an effective plant-derived antifungal formulation that can provide an alternative to currently available antifungal drugs.


Assuntos
Compostos Alílicos/uso terapêutico , Antifúngicos/uso terapêutico , Candidíase/tratamento farmacológico , Sulfetos/uso terapêutico , Compostos Alílicos/administração & dosagem , Animais , Antifúngicos/administração & dosagem , Candida albicans/efeitos dos fármacos , Feminino , Lipossomos , Camundongos , Camundongos Endogâmicos BALB C , Modelos Animais , Sulfetos/administração & dosagem
18.
J Drug Target ; 18(1): 45-52, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19624287

RESUMO

The present study deals with the evaluation of the efficacy of amoxicillin bearing poly-lactic-glycolic acid (PLGA) microsphere formulation in treatment of experimental listeriosis in Swiss albino mice. Amoxicillin bearing PLGA microspheres were prepared by water-in-oil-in-water emulsion technique. PLGA microwspheres significantly regulated sustained release of encapsulated drug over extended time period. The rate of release increased in temperature dependent manner. Amoxicillin bearing PLGA microsphere successfully cleared bacterial burdens in vital organs (kidney, spleen, and brain) and also increased survival rate of treated animals in comparison to free form of the drug. The higher efficacy of microsphere based novel formulation of amoxicillin could be attributed to its targeted delivery to infected macrophages as well as sustained release over extended period of time.


Assuntos
Amoxicilina/administração & dosagem , Antibacterianos/administração & dosagem , Ácido Láctico/química , Listeriose/tratamento farmacológico , Ácido Poliglicólico/química , Amoxicilina/farmacologia , Animais , Antibacterianos/farmacologia , Química Farmacêutica , Preparações de Ação Retardada , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos , Feminino , Listeria monocytogenes/isolamento & purificação , Listeriose/microbiologia , Listeriose/mortalidade , Macrófagos/metabolismo , Camundongos , Microesferas , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Taxa de Sobrevida , Temperatura , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...