Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Adv Sci (Weinh) ; : e2310204, 2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-38937984

RESUMO

The development of immune cell engagers (ICEs) can be limited by logistical and functional restrictions associated with fusion protein designs, thus limiting immune cell recruitment to solid tumors. Herein, a high affinity superantigen-based multivalent ICE is developed for simultaneous activation and recruitment of NK and T cells for tumor treatment. Yeast library-based directed evolution is adopted to identify superantigen variants possessing enhanced binding affinity to immunoreceptors expressed on human T cells and NK cells. High-affinity superantigens exhibiting improved immune-stimulatory activities are then incorporated into a superantigen-based tri-functional yeast-display-enhanced multivalent immune cell engager (STYMIE), which is functionalized with a nanobody, a Neo-2/15 cytokine, and an Fc domain for tumor targeting, immune stimulation, and prolonged circulation, respectively. Intravenous administration of STYMIE enhances NK and T cell recruitment into solid tumors, leading to enhanced inhibition in multiple tumor models. The study offers design principles for multifunctional ICEs.

2.
Elife ; 122024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38747577

RESUMO

Certain bacteria demonstrate the ability to target and colonize the tumor microenvironment, a characteristic that positions them as innovative carriers for delivering various therapeutic agents in cancer therapy. Nevertheless, our understanding of how bacteria adapt their physiological condition to the tumor microenvironment remains elusive. In this work, we employed liquid chromatography-tandem mass spectrometry to examine the proteome of E. coli colonized in murine tumors. Compared to E. coli cultivated in the rich medium, we found that E. coli colonized in tumors notably upregulated the processes related to ferric ions, including the enterobactin biosynthesis and iron homeostasis. This finding indicated that the tumor is an iron-deficient environment to E. coli. We also found that the colonization of E. coli in the tumor led to an increased expression of lipocalin 2 (LCN2), a host protein that can sequester the enterobactin. We therefore engineered E. coli in order to evade the nutritional immunity provided by LCN2. By introducing the IroA cluster, the E. coli synthesizes the glycosylated enterobactin, which creates steric hindrance to avoid the LCN2 sequestration. The IroA-E. coli showed enhanced resistance to LCN2 and significantly improved the anti-tumor activity in mice. Moreover, the mice cured by the IroA-E. coli treatment became resistant to the tumor re-challenge, indicating the establishment of immunological memory. Overall, our study underscores the crucial role of bacteria's ability to acquire ferric ions within the tumor microenvironment for effective cancer therapy.


Assuntos
Escherichia coli , Ferro , Lipocalina-2 , Animais , Escherichia coli/genética , Escherichia coli/metabolismo , Lipocalina-2/metabolismo , Lipocalina-2/genética , Camundongos , Ferro/metabolismo , Neoplasias/terapia , Neoplasias/imunologia , Enterobactina/metabolismo , Microambiente Tumoral , Linhagem Celular Tumoral
3.
EMBO Mol Med ; 16(2): 416-428, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38225455

RESUMO

The tumor microenvironment (TME) presents differential selective pressure (DSP) that favors the growth of cancer cells, and monovalent therapy is often inadequate in reversing the cancer cell dominance in the TME. In this work, we introduce bacteria as a foreign species to the TME and explore combinatorial treatment strategies to alter DSP for tumor eradication. We show that cancer-selective chemotherapeutic agents and fasting can provide a strong selection pressure against tumor growth in the presence of bacteria. Moreover, we show that an immunogenic drug (oxaliplatin), but not a non-immunogenic one (5-FU), synergizes with the bacteria to activate both the innate and adaptive immunity in the TME, resulting in complete tumor remission and a sustained anti-tumor immunological memory in mice. The combination of oxaliplatin and bacteria greatly enhances the co-stimulatory and antigen-presenting molecules on antigen-presenting cells, which in turn bridge the cytotoxic T cells for cancer-cell killing. Our findings indicate that rational combination of bacterial therapy and immunogenic chemotherapy can promote anticancer immunity against the immunosuppressive TME.


Assuntos
Antineoplásicos , Neoplasias , Animais , Camundongos , Oxaliplatina/uso terapêutico , Microambiente Tumoral , Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Linfócitos T Citotóxicos , Imunoterapia/métodos , Linhagem Celular Tumoral
4.
Trends Biotechnol ; 42(2): 241-252, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-37743158

RESUMO

An emerging cellular engineering method creates synthetic polymer matrices inside cells. By contrast with classical genetic, enzymatic, or radioactive techniques, this materials-based approach introduces non-natural polymers inside cells, thus modifying cellular states and functionalities. Here, we cover various materials and chemistries that have been exploited to create intracellular polymer matrices. In addition, we discuss emergent cellular properties due to the intracellular polymerization, including nonreplicating but active metabolism, maintenance of membrane integrity, and resistance to environmental stressors. We also discuss past work and future opportunities for developing and applying synthetic cells that contain intracellular polymers. The materials-based approach will usher in new applications of synthetic cells for broad biotechnological applications.


Assuntos
Biotecnologia , Polímeros , Polimerização , Engenharia Celular , Materiais Biocompatíveis
5.
Adv Healthc Mater ; 12(8): e2201708, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36455286

RESUMO

The intricate functionalities of cellular membranes have inspired strategies for deriving and anchoring cell-surface components onto solid substrates for biological studies, biosensor applications, and tissue engineering. However, introducing conformal and right-side-out cell membrane coverage onto planar substrates requires cumbersome protocols susceptible to significant device-to-device variability. Here, a facile approach for biomembrane functionalization of planar substrates is demonstrated by subjecting confluent cellular monolayer to intracellular hydrogel polymerization. The resulting cell-gel hybrid, herein termed GELL (gelated cell), exhibits extraordinary stability and retains the structural integrity, membrane fluidity, membrane protein mobility, and topology of living cells. In assessing the utility of GELL layers as a tissue engineering feeder substrate for stem cell maintenance, GELL feeder prepared from primary mouse embryonic fibroblasts not only preserves the stemness of murine stem cells but also exhibits advantages over live feeder cells owing to the GELL's inanimate, non-metabolizing nature. The preparation of a xeno-free feeder substrate devoid of non-human components is further shown with HeLa cells, and the resulting  HeLa GELL feeder effectively sustains the growth and stemness of both murine and human induced pluripotent stem cells. The study highlights a novel bio-functionalization strategy that introduces new opportunities for tissue engineering and other biomedical applications.


Assuntos
Células-Tronco Pluripotentes Induzidas , Células-Tronco Pluripotentes , Humanos , Animais , Camundongos , Fibroblastos , Células HeLa , Células Alimentadoras/metabolismo , Diferenciação Celular
6.
Adv Sci (Weinh) ; 9(13): e2105506, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35246961

RESUMO

Membrane-lytic peptides offer broad synthetic flexibilities and design potential to the arsenal of anticancer therapeutics, which can be limited by cytotoxicity to noncancerous cells and induction of drug resistance via stress-induced mutagenesis. Despite continued research efforts on membrane-perforating peptides for antimicrobial applications, success in anticancer peptide therapeutics remains elusive given the muted distinction between cancerous and normal cell membranes and the challenge of peptide degradation and neutralization upon intravenous delivery. Using triple-negative breast cancer as a model, the authors report the development of a new class of anticancer peptides. Through function-conserving mutations, the authors achieved cancer cell selective membrane perforation, with leads exhibiting a 200-fold selectivity over non-cancerogenic cells and superior cytotoxicity over doxorubicin against breast cancer tumorspheres. Upon continuous exposure to the anticancer peptides at growth-arresting concentrations, cancer cells do not exhibit resistance phenotype, frequently observed under chemotherapeutic treatment. The authors further demonstrate efficient encapsulation of the anticancer peptides in 20 nm polymeric nanocarriers, which possess high tolerability and lead to effective tumor growth inhibition in a mouse model of MDA-MB-231 triple-negative breast cancer. This work demonstrates a multidisciplinary approach for enabling translationally relevant membrane-lytic peptides in oncology, opening up a vast chemical repertoire to the arms race against cancer.


Assuntos
Antineoplásicos , Neoplasias de Mama Triplo Negativas , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Humanos , Camundongos , Peptídeos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/metabolismo
7.
Mol Ther Methods Clin Dev ; 21: 299-314, 2021 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-33898629

RESUMO

Antigen-specific lung-resident memory T cells (TRMs) constitute the first line of defense that mediates rapid protection against respiratory pathogens and inspires novel vaccine designs against infectious pandemic threats, yet effective means of inducing TRMs, particularly via non-viral vectors, remain challenging. Here, we demonstrate safe and potent induction of lung-resident TRMs using a biodegradable polymeric nanoshell that co-encapsulates antigenic peptides and TLR9 agonist CpG-oligodeoxynucleotide (CpG-ODN) in a virus-mimicking structure. Through subcutaneous priming and intranasal boosting, the combinatorial nanoshell vaccine elicits prominent lung-resident CD4+ and CD8+ T cells that surprisingly show better durability than live viral infections. In particular, nanoshells containing CpG-ODN and a pair of conserved class I and II major histocompatibility complex-restricted influenza nucleoprotein-derived antigenic peptides are demonstrated to induce near-sterilizing immunity against lethal infections with influenza A viruses of different strains and subtypes in mice, resulting in rapid elimination of replicating viruses. We further examine the pulmonary transport dynamic and optimal composition of the nanoshell vaccine conducive for robust TRM induction as well as the benefit of subcutaneous priming on TRM replenishment. The study presents a practical vaccination strategy for inducing protective TRM-mediated immunity, offering a compelling platform and critical insights in the ongoing quest toward a broadly protective vaccine against universal influenza as well as other respiratory pathogens.

8.
Drug Deliv Transl Res ; 11(4): 1420-1437, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33748879

RESUMO

The COVID-19 pandemic's high mortality rate and severe socioeconomic impact serve as a reminder of the urgent need for effective countermeasures against viral pandemic threats. In particular, effective antiviral therapeutics capable of stopping infections in its tracks is critical to reducing infection fatality rate and healthcare burden. With the field of drug delivery witnessing tremendous advancement in the last two decades owing to a panoply of nanotechnology advances, the present review summarizes and expounds on the research and development of therapeutic nanoformulations against various infectious viral pathogens, including HIV, influenza, and coronaviruses. Specifically, nanotechnology advances towards improving pathogen- and host-targeted antiviral drug delivery are reviewed, and the prospect of achieving effective viral eradication, broad-spectrum antiviral effect, and resisting viral mutations are discussed. As several COVID-19 antiviral clinical trials are met with lackluster treatment efficacy, nanocarrier strategies aimed at improving drug pharmacokinetics, biodistributions, and synergism are expected to not only contribute to the current disease treatment efforts but also expand the antiviral arsenal against other emerging viral diseases.


Assuntos
Antivirais/administração & dosagem , COVID-19/prevenção & controle , Sistemas de Liberação de Medicamentos/métodos , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Nanopartículas/administração & dosagem , Nanotecnologia/métodos , Animais , Antivirais/imunologia , COVID-19/epidemiologia , COVID-19/imunologia , Sistemas de Liberação de Medicamentos/tendências , Interações Hospedeiro-Patógeno/imunologia , Humanos , Nanotecnologia/tendências , Pandemias/prevenção & controle , Viroses/epidemiologia , Viroses/imunologia , Viroses/prevenção & controle , Replicação Viral/efeitos dos fármacos , Replicação Viral/fisiologia
9.
Hum Vaccin Immunother ; 17(3): 654-655, 2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-32991231

RESUMO

A safe and effective vaccine candidate is urgently needed for the ongoing COVID-19 pandemic, caused by SARS-CoV-2. Here we report that recombinant SARS-CoV-2 RBD protein immunization in mice is able to elicit a strong antibody response and potent neutralizing capability as measured using live or pseudotyped SARS-CoV-2 neutralization assays.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , COVID-19/imunologia , Ligação Proteica/imunologia , Domínios Proteicos/imunologia , SARS-CoV-2/imunologia , Animais , Linhagem Celular , Células HEK293 , Humanos , Camundongos , Pandemias/prevenção & controle , Proteínas Recombinantes/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia
10.
Nanomedicine (Lond) ; 15(29): 2883-2894, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33252301

RESUMO

The discovery of stimulator of interferon genes (STING) and their agonists as primary components that link antiviral innate and adaptive immunity has motivated growing research on STING agonist-mediated immunotherapy and vaccine development. To overcome the delivery challenge in shuttling highly polar STING agonists, typically in the form of cyclic dinucleotides, to target cells and to STING proteins in cellular cytosol, numerous nanoformulation strategies have been implemented for effective STING activation. While many STING-activating nanoparticles are developed to enhance anticancer immunotherapy, their adoption as vaccine adjuvant has vastly propelled antiviral vaccination efforts against challenging public health threats, including HIV, influenza and coronaviruses. In light of the COVID-19 pandemic that has thrusted vaccine development into the public spotlight, this review highlights advances in nanomedicinal STING agonist delivery with an emphasis on their applications in antiviral vaccination.


Assuntos
Tratamento Farmacológico da COVID-19 , Vacinas contra COVID-19/uso terapêutico , Imunidade Inata/efeitos dos fármacos , Pandemias , Antivirais/uso terapêutico , COVID-19/patologia , COVID-19/virologia , Vacinas contra COVID-19/imunologia , Humanos , Imunoterapia/tendências , Nanopartículas/química , Nanopartículas/uso terapêutico , SARS-CoV-2/imunologia , SARS-CoV-2/patogenicidade , Transdução de Sinais/efeitos dos fármacos
11.
Int J Nanomedicine ; 15: 3303-3318, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32494131

RESUMO

BACKGROUND: Poultry vaccine has limited choices of adjuvants and is facing severe threat of infectious diseases due to ineffective of widely used commercial vaccines. Thus, development of novel adjuvant that offers safe and effective immunity is of urgent need. MATERIALS AND METHODS: The present research engineers a novel chicken adjuvant with potent immune-potentiating capability by incorporating avian toll-like receptor 21 (TLR21) agonist CpG ODN 2007 with a poly(lactic-co-glycolic acid) (PLGA)-based hollow nanoparticle platform (CpG-NP), which subsequently assessed ex vivo and in vivo. RESULTS: CpG-NPs with an average diameter of 164 nm capable of sustained release of CpG for up to 96 hours were successfully prepared. With the ex vivo model of chicken bone marrow-derived dendritic cells (chBMDCs), CpG-NP was engulfed effectively and found to induce DC maturation, promoting dendrite formation and upregulation of CD40, CD80 and CCR7. In addition to enhanced expression of IL-1ß, IL-6, IL-12 and IFN-γ, 53/84 immune-related genes were found to be stimulated in CpG-NP-treated chBMDCs, whereas only 39 of such genes were stimulated in free CpG-treated cells. These upregulated genes suggest immune skewing toward T helper cell 1 bias and evidence of improved mucosal immunity upon vaccination with the CpG-NP. The CpG-NP-treated chBMDCs showed protective effects to DF-1 cells against avian influenza virus H6N1 infection. Upon subsequent coupling with infectious bronchitis virus subunit antigen administration, chickens were immunostimulated to acquire higher humoral immune response and protective response against viral challenge. CONCLUSTION: This work presents a novel hollow CpG-NP formulation, demonstrating effective and long-lasting immunostimulatory ability ex vivo and in vivo for chickens, as systemically compared to free CpG. This enhanced immune stimulation benefits from high stability and controlled release of internal component of nanoparticles that improve cellular delivery, lymphoid organ targeting and sustainable DC activation. CpG-NP has broad application potential in antiviral and vaccine development.


Assuntos
Antivirais/farmacologia , Galinhas/imunologia , Imunidade/efeitos dos fármacos , Nanopartículas/química , Oligodesoxirribonucleotídeos/farmacologia , Polímeros/química , Vacinas/imunologia , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Cães , Imunidade Humoral/efeitos dos fármacos , Imunização , Vírus da Bronquite Infecciosa/efeitos dos fármacos , Células Madin Darby de Rim Canino , Nanopartículas/administração & dosagem , Nanopartículas/ultraestrutura , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química
12.
Nano Lett ; 20(4): 2246-2256, 2020 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-32160474

RESUMO

Many favorable anticancer treatments owe their success to the induction immunogenic cell death (ICD) in cancer cells, which results in the release of endogenous danger signals along with tumor antigens for effective priming of anticancer immunity. We describe a strategy to artificially induce ICD by delivering the agonist of stimulator of interferon genes (STING) into tumor cells using hollow polymeric nanoshells. Following intracellular delivery of exogenous adjuvant, subsequent cytotoxic treatment creates immunogenic cellular debris that spatiotemporally coordinate tumor antigens and STING agonist in a process herein termed synthetic immunogenic cell death (sICD). sICD is indiscriminate to the type of chemotherapeutics and enables colocalization of exogenously administered immunologic adjuvants and tumor antigens for enhanced antigen presentation and anticancer adaptive response. In three mouse tumor models, sICD enhances therapeutic efficacy and restrains tumor progression. The study highlights the benefit of delivering STING agonists to cancer cells, paving ways to new chemo-immunotherapeutic designs.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Morte Celular Imunogênica/efeitos dos fármacos , Proteínas de Membrana/agonistas , Nanoconchas/uso terapêutico , Neoplasias/terapia , Animais , Antineoplásicos Imunológicos/administração & dosagem , Linhagem Celular Tumoral , Progressão da Doença , Humanos , Imunoterapia , Camundongos Endogâmicos BALB C , Nanoconchas/administração & dosagem , Neoplasias/imunologia
13.
Adv Funct Mater ; 29(28): 1807616, 2019 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-32313544

RESUMO

The continued threat of emerging, highly lethal infectious pathogens such as Middle East respiratory syndrome coronavirus (MERS-CoV) calls for the development of novel vaccine technology that offers safe and effective prophylactic measures. Here, a novel nanoparticle vaccine is developed to deliver subunit viral antigens and STING agonists in a virus-like fashion. STING agonists are first encapsulated into capsid-like hollow polymeric nanoparticles, which show multiple favorable attributes, including a pH-responsive release profile, prominent local immune activation, and reduced systemic reactogenicity. Upon subsequent antigen conjugation, the nanoparticles carry morphological semblance to native virions and facilitate codelivery of antigens and STING agonists to draining lymph nodes and immune cells for immune potentiation. Nanoparticle vaccine effectiveness is supported by the elicitation of potent neutralization antibody and antigen-specific T cell responses in mice immunized with a MERS-CoV nanoparticle vaccine candidate. Using a MERS-CoV-permissive transgenic mouse model, it is shown that mice immunized with this nanoparticle-based MERS-CoV vaccine are protected against a lethal challenge of MERS-CoV without triggering undesirable eosinophilic immunopathology. Together, the biocompatible hollow nanoparticle described herein provides an excellent strategy for delivering both subunit vaccine candidates and novel adjuvants, enabling accelerated development of effective and safe vaccines against emerging viral pathogens.

14.
Int J Nanomedicine ; 13: 8579-8593, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30587980

RESUMO

BACKGROUND: Influenza virus infections are a major public health concern worldwide. Conventional treatments against the disease are designed to target viral proteins. However, the emergence of viral variants carrying drug-resistant mutations can outpace the development of pathogen-targeting antivirals. Diphyllin and bafilomycin are potent vacuolar ATPase (V-ATPase) inhibitors previously shown to have broad-spectrum antiviral activity. However, their poor water solubility and potential off-target effect limit their clinical application. METHODS: In this study, we report that nanoparticle encapsulation of diphyllin and bafilomycin improves the drugs' anti-influenza applicability. RESULTS: Using PEG-PLGA diblock copolymers, sub-200 nm diphyllin and bafilomycin nanoparticles were prepared, with encapsulation efficiency of 42% and 100%, respectively. The drug-loaded nanoparticles have sustained drug release kinetics beyond 72 hours and facilitate intracellular drug delivery to two different influenza virus-permissive cell lines. As compared to free drugs, the nanoparticulate V-ATPase inhibitors exhibited lower cytotoxicity and greater in vitro antiviral activity, improving the therapeutic index of diphyllin and bafilomycin by approximately 3 and 5-fold, respectively. In a mouse model of sublethal influenza challenge, treatment with diphyllin nanoparticles resulted in reduced body weight loss and viral titer in the lungs. In addition, following a lethal influenza viral challenge, diphyllin nanoparticle treatment conferred a survival advantage of 33%. CONCLUSIONS: These results demonstrate the potential of the nanoparticulate V-ATPase inhibitors for host-targeted treatment against influenza.


Assuntos
Antivirais/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Influenza Humana/tratamento farmacológico , Nanopartículas/química , ATPases Vacuolares Próton-Translocadoras/antagonistas & inibidores , Animais , Antivirais/química , Antivirais/farmacologia , Benzodioxóis/química , Benzodioxóis/farmacologia , Benzodioxóis/uso terapêutico , Linhagem Celular , Cães , Liberação Controlada de Fármacos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Humanos , Influenza Humana/virologia , Concentração Inibidora 50 , Cinética , Lignanas/química , Lignanas/farmacologia , Lignanas/uso terapêutico , Macrolídeos/química , Macrolídeos/farmacologia , Macrolídeos/uso terapêutico , Camundongos , Nanopartículas/ultraestrutura , Orthomyxoviridae/efeitos dos fármacos , Orthomyxoviridae/fisiologia , Infecções por Orthomyxoviridae/tratamento farmacológico , Infecções por Orthomyxoviridae/virologia , ATPases Vacuolares Próton-Translocadoras/metabolismo , Proteínas Virais/metabolismo , Replicação Viral/efeitos dos fármacos
15.
Acta Biomater ; 82: 133-142, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30316023

RESUMO

Amidst the ever-rising threat of antibiotics resistance, colistin, a decade-old antibiotic with lingering toxicity concern, is increasingly prescribed to treat many drug-resistant, gram-negative bacteria. With the aim of improving the safety profile while preserving the antimicrobial activity of colistin, a nanoformulation is herein developed through coacervate complexation with polyanionic peptides. Upon controlled mixing of cationic colistin with polyglutamic acids, formation of liquid coacervates was demonstrated. Subsequent stabilization by DSPE-PEG and homogenization through micro-fluidization of the liquid coacervates yielded nanoparticles 8 nm in diameter. In vitro assessment showed that the colistin antimicrobial activity against multiple drug-resistant bacterial strains was retained and, in some cases, enhanced following the nanoparticle assembly. In vivo administration in mice demonstrated improved safety of the colistin nanoparticle, which has a maximal tolerated dose of 12.5 mg/kg compared to 10 mg/kg of free colistin. Upon administration over a 7-day period, colistin nanoparticles also exhibited reduced hepatotoxicity as compared to free colistin. In mouse models of Klebsiella pneumoniae bacteremia and Acinetobacter baumannii pneumonia, treatment with colistin nanoparticles showed equivalent efficacy to free colistin. These results demonstrate coacervation-induced nanoparticle assembly as a promising approach towards improving colistin treatments against bacterial infections. STATEMENT OF SIGNIFICANCE: Improving the safety of colistin while retaining its antimicrobial activity has been a highly sought-after objective toward enhancing antibacterial treatments. Herein, we demonstrate formation of stabilized colistin nanocomplexes in the presence of anionic polypeptides and DSPE-PEG stabilizer. The nanocomplexes retain colistin's antimicrobial activity while demonstrating improved safety upon in vivo administration. The supramolecular nanoparticle assembly of colistin presents a unique approach towards designing antimicrobial nanoparticles.


Assuntos
Infecções por Acinetobacter , Acinetobacter baumannii/metabolismo , Bacteriemia , Colistina , Infecções por Klebsiella , Klebsiella pneumoniae/metabolismo , Nanopartículas , Pneumonia Bacteriana , Infecções por Acinetobacter/tratamento farmacológico , Infecções por Acinetobacter/metabolismo , Animais , Bacteriemia/tratamento farmacológico , Bacteriemia/metabolismo , Colistina/química , Colistina/farmacologia , Infecções por Klebsiella/tratamento farmacológico , Infecções por Klebsiella/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas/química , Nanopartículas/uso terapêutico , Pneumonia Bacteriana/tratamento farmacológico , Pneumonia Bacteriana/metabolismo
16.
Adv Healthc Mater ; 7(13): e1701395, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29508547

RESUMO

As the dawn of the postantibiotic era we approach, antibacterial vaccines are becoming increasingly important for managing bacterial infection and reducing the need for antibiotics. Despite the success of vaccination, vaccines remain unavailable for many pressing microbial diseases, including tuberculosis, chlamydia, and staphylococcus infections. Amid continuing research efforts in antibacterial vaccine development, the advancement of nanomaterial engineering has brought forth new opportunities in vaccine designs. With increasing knowledge in antibacterial immunity and immunologic adjuvants, innovative nanoparticles are designed to elicit the appropriate immune responses for effective antimicrobial defense. Rationally designed nanoparticles are demonstrated to overcome delivery barriers to shape the adaptive immunity. This article reviews the advances in nanoparticle- and nanomaterial-based antibacterial vaccines and summarizes the development of nanoparticulate adjuvants for immune potentiation against microbial pathogens. In addition, challenges and progress in ongoing antibacterial vaccine development are discussed to highlight the opportunities for future vaccine designs.


Assuntos
Infecções Bacterianas/prevenção & controle , Vacinas Bacterianas/farmacologia , Nanoestruturas/química , Receptores de Reconhecimento de Padrão/agonistas , Vacinas Bacterianas/imunologia , Humanos , Nanopartículas/química , Receptores de Reconhecimento de Padrão/imunologia
17.
Nanotheranostics ; 1(3): 244-260, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29071191

RESUMO

Synthetic nanoparticles play an increasingly significant role in vaccine design and development as many nanoparticle vaccines show improved safety and efficacy over conventional formulations. These nanoformulations are structurally similar to viruses, which are nanoscale pathogenic organisms that have served as a key selective pressure driving the evolution of our immune system. As a result, mechanisms behind the benefits of nanoparticle vaccines can often find analogue to the interaction dynamics between the immune system and viruses. This review covers the advances in vaccine nanotechnology with a perspective on the advantages of virus mimicry towards immune potentiation. It provides an overview to the different types of nanomaterials utilized for nanoparticle vaccine development, including functionalization strategies that bestow nanoparticles with virus-like features. As understanding of human immunity and vaccine mechanisms continue to evolve, recognizing the fundamental semblance between synthetic nanoparticles and viruses may offer an explanation for the superiority of nanoparticle vaccines over conventional vaccines and may spur new design rationales for future vaccine research. These nanoformulations are poised to provide solutions towards pressing and emerging human diseases.

18.
Sci Rep ; 7(1): 13043, 2017 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-29026122

RESUMO

Feline infectious peritonitis (FIP), caused by a mutated feline coronavirus, is one of the most serious and fatal viral diseases in cats. The disease remains incurable, and there is no effective vaccine available. In light of the pathogenic mechanism of feline coronavirus that relies on endosomal acidification for cytoplasmic entry, a novel vacuolar ATPase blocker, diphyllin, and its nanoformulation are herein investigated for their antiviral activity against the type II feline infectious peritonitis virus (FIPV). Experimental results show that diphyllin dose-dependently inhibits endosomal acidification in fcwf-4 cells, alters the cellular susceptibility to FIPV, and inhibits the downstream virus replication. In addition, diphyllin delivered by polymeric nanoparticles consisting of poly(ethylene glycol)-block-poly(lactide-co-glycolide) (PEG-PLGA) further demonstrates an improved safety profile and enhanced inhibitory activity against FIPV. In an in vitro model of antibody-dependent enhancement of FIPV infection, diphyllin nanoparticles showed a prominent antiviral effect against the feline coronavirus. In addition, the diphyllin nanoparticles were well tolerated in mice following high-dose intravenous administration. This study highlights the therapeutic potential of diphyllin and its nanoformulation for the treatment of FIP.


Assuntos
Antivirais/farmacologia , Coronavirus Felino/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Nanopartículas/química , ATPases Vacuolares Próton-Translocadoras/antagonistas & inibidores , Animais , Benzodioxóis/farmacologia , Gatos , Linhagem Celular , Modelos Animais de Doenças , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Peritonite Infecciosa Felina/imunologia , Peritonite Infecciosa Felina/virologia , Lignanas/farmacologia , Nanopartículas/ultraestrutura , Polietilenoglicóis/química , ATPases Vacuolares Próton-Translocadoras/metabolismo
19.
BMC Biotechnol ; 17(1): 2, 2017 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-28061848

RESUMO

BACKGROUND: Human infection with avian influenza A virus (H7N9) was first reported in China in March 2013. Since then, hundreds of cases have been confirmed showing severe symptoms with a high mortality rate. The virus was transmitted from avian species to humans and has spread to many neighboring areas, raising serious concerns over its pandemic potential. Towards containing the disease, the goal of this study is to prepare a virus-like particle (VLP) that consists of hemagglutinin (HA), neuraminidase (NA) and matrix protein 1 (M1) derived from the human isolate A/Taiwan/S02076/2013(H7N9) for potential vaccine development. RESULTS: Full length HA, NA, and M1 protein genes were cloned and expressed using a baculoviral expression system, and the VLPs were generated by co-infecting insect cells with three respective recombinant baculoviruses. Nanoparticle tracking analysis and transmission electron microscopy were applied to verify the VLPs' structure and antigenicity, and the multiplicity of infection of the recombinant baculoviruses was adjusted to achieve the highest hemagglutination activity. In animal experiments, BALB/c mice and specific-pathogen-free chickens receiving the VLP immunization showed elevated hemagglutination inhibition serum titer and antibodies against NA and M1 proteins. In addition, examination of cellular immunity showed the VLP-immunized mice and chickens exhibited an increased splenic antigen-specific cytokines production. CONCLUSIONS: The H7N9 VLPs possess desirable immunogenicity in vivo and may serve as a candidate for vaccine development against avian influenza A (H7N9) infection.


Assuntos
Antígenos Virais/imunologia , Galinhas/imunologia , Vírus da Influenza A/imunologia , Camundongos/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Animais , Antígenos Virais/genética , Feminino , Vírus da Influenza A/genética , Camundongos Endogâmicos BALB C , Engenharia de Proteínas , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Especificidade da Espécie , Vacinas de Partículas Semelhantes a Vírus/genética
20.
Nanoscale ; 6(5): 2730-7, 2014 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-24463706

RESUMO

The unique structural features and stealth properties of a recently developed red blood cell membrane-cloaked nanoparticle (RBC-NP) platform raise curiosity over the interfacial interactions between natural cellular membranes and polymeric nanoparticle substrates. Herein, several interfacial aspects of the RBC-NPs are examined, including completeness of membrane coverage, membrane sidedness upon coating, and the effects of polymeric particles' surface charge and surface curvature on the membrane cloaking process. The study shows that RBC membranes completely cover negatively charged polymeric nanoparticles in a right-side-out manner and enhance the particles' colloidal stability. The membrane cloaking process is applicable to particle substrates with a diameter ranging from 65 to 340 nm. Additionally, the study reveals that both surface glycans on RBC membranes and the substrate properties play a significant role in driving and directing the membrane-particle assembly. These findings further the understanding of the dynamics between cellular membranes and nanoscale substrates and provide valuable information toward future development and characterization of cellular membrane-cloaked nanodevices.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA