Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biomed Pharmacother ; 155: 113682, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36095964

RESUMO

Gram-negative, facultatively anaerobic bacteria Salmonella Typhimurium is a candidate agent or delivery vector for cancer therapy. Effective targeted therapies in addition to radiotherapy, chemotherapy and surgery have been urgently needed as an alternative or supplement. This study expected to further improve the tumor-targeting ability of Salmonella bacteria through genetic modifications. Based on an auxotrophic Salmonella bacterial strain (D2), we constructed Salmonella mutants with altered LPS length to facilitate displaying the RGD4C targeting peptide on the outer membrane surface of Salmonella. The expression of RGD4C peptide in fusion with OmpA was identified by outer membrane protein extraction and WB detection in different mutant strains. However, flow cytometry analysis following immunofluorescence staining demonstrated that the extracellular length of Salmonella LPS did affect the surface display of RGD4C peptide. The strain D2-RGD4C that synthesized intact LPS including lipid A, core oligosaccharides and O antigen polysaccharides could hardly display RGD4C peptide, showing the same fluorescence signal intensity as the strains not expressing RGD4C peptide. Among different strains, D2 ∆rfaJ-RGD4C that synthesized truncated LPS including lipid A and partial core oligosaccharides was capable of displaying RGD4C peptide most efficiently and showed the highest ability to target HUVECs expressing αV integrin and tumor tissue with abundant neovascularization. Animal experiments also demonstrated that this tumor-targeting attenuated Salmonella strain to simultaneously deliver endostatin and TRAIL, two agents with different anti-tumor activities, could significantly inhibit tumor growth and prolong mouse survival. Thus, our studies revealed that Salmonella could be genetically engineered to improve its tumor targeting via the truncation of LPS and surface display of targeting peptides, thereby eliciting superior anti-tumor effects through targeted delivery of drug molecules.


Assuntos
Neoplasias , Salmonella typhimurium , Camundongos , Animais , Antígenos O/metabolismo , Lipopolissacarídeos/farmacologia , Endostatinas/farmacologia , Lipídeo A/metabolismo , Lipídeo A/farmacologia , Integrina alfaV/metabolismo , Peptídeos/farmacologia , Peptídeos/metabolismo
2.
Front Microbiol ; 12: 774490, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35003007

RESUMO

The gram-negative facultative anaerobic bacteria Salmonella enterica serovar Typhimurium (hereafter S. Typhimurium) has always been considered as one candidate of anti-tumor agents or vectors for delivering drug molecules. In this study, we compared several widely studied S. Typhimurium strains in their anti-tumor properties aiming to screen out the best one for further optimization and use in cancer therapy. In terms of the motility, virulence and anti-tumor efficacy, the three strains 14028, SL1344, and UK-1 were similar and obviously better than LT-2, and UK-1 showed the best phenotypes among them. Therefore, the strain UK-1 (D) was selected for the following studies. Its auxotrophic mutant strain (D1) harboring ∆aroA and ∆purM mutations was further optimized through the modification of lipid A structure, generating a new strain named D2 with stronger immunostimulatory activity. Finally, the ∆asd derivative of D2 was utilized as one live vector to deliver anti-tumor molecules including the angiogenesis inhibitor endostatin and apoptosis inducer TRAIL and the therapeutic and toxic-side effects were evaluated in mouse models of colon carcinoma and melanoma. After intraperitoneal infection, engineered Salmonella bacteria equipped with endostatin and/or TRAIL significantly suppressed the tumor growth and prolonged survival of tumor-bearing mice compared to PBS or bacteria carrying the empty plasmid. Consistently, immunohistochemical studies confirmed the colonization of Salmonella bacteria and the expression of anti-tumor molecules inside tumor tissue, which were accompanied by the increase of cell apoptosis and suppression of tumor angiogenesis. These results demonstrated that the beneficial anti-tumor efficacy of attenuated S. Typhimurium bacteria could be improved through delivery of drug molecules with powerful anti-tumor activities.

3.
Biotechnol Bioeng ; 118(2): 513-530, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33038015

RESUMO

Cancer has always been a global problem, with more cases of cancer patients being diagnosed every year. Conventional cancer treatments, including radiotherapy, chemotherapy, and surgery, are still unable to bypass their obvious limitations, and developing effective targeted therapies is still required. More than one century ago, the doctor William B. Coley discovered that cancer patients had tumor regression by injection of Streptococcus bacteria. The studies of cancer therapy using bacterial microorganisms are now very widespread. In particular, the facultative anaerobic bacteria Salmonella typhimurium is widely investigated as it can selectively colonize different types of tumors, locally deliver various antitumor drugs, and inhibit tumor growth. The exciting antitumor efficacy and safety observed in animal tumor models prompted the well-known attenuated Salmonella bacterial strain VNP20009 to be tested in human clinical trials in the early 21st century. Regrettably, no patients showed significant therapeutic effects and even bacterial colonization in tumor tissue was undetectable in most patients. Salmonella bacteria are still considered as a promising agent or vehicle for cancer therapy. Recent efforts have been focused on the generation of attenuated bacterial strains with higher targeting for tumor tissue, and optimization of the delivery of therapeutic antitumor cargoes into the tumor microenvironment. This review will summarize new technologies or approaches that may improve bacteria-mediated cancer therapy.


Assuntos
Microrganismos Geneticamente Modificados , Neoplasias/terapia , Salmonella typhimurium , Microambiente Tumoral , Animais , Humanos
4.
Cancer Lett ; 491: 11-21, 2020 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-32721550

RESUMO

Bacteria are always a considerable tool for the cancer therapy. The bacteria-derived minicells are re-emerged as a promising drug delivery system for cancer therapy. The minicells are nano-sized, anucleated, non-dividing, and metabolically active cells produced by abnormal bacterial cell division that are able to transcribe and translate the gene of interest. Minicells encapsulate a wide range of chemotherapeutic and molecular drugs, si/shRNA, antigens and therapeutic toxins to precisely deliver them to the cancer cells through the easy modification of minicell surface with bi-specific antibodies against receptor-targeted cancer cells. Minicell-mediated chemotherapy may inhibit the growth of drug-resistant tumors and exhibits the potential to successfully deliver the chemotherapeutics into hypoxic and necrotic region in solid tumors. This novel approach significantly overcomes drug leakage and severe toxicity by enhancing targeting specificity and therapeutic index of drugs in cancer therapy. Many antibody-conjugated drug-loaded minicells are being investigated in clinical trials for cancer therapy. This review summarizes the advantages of bacteria-derived minicells as delivery systems for anti-cancer drugs or agents and the recent advances and emerging future in cancer therapy.


Assuntos
Antineoplásicos/administração & dosagem , Bactérias/genética , Sistemas de Liberação de Medicamentos , Neoplasias/tratamento farmacológico , Humanos , Nanotecnologia
5.
Sheng Wu Gong Cheng Xue Bao ; 35(6): 998-1008, 2019 Jun 25.
Artigo em Chinês | MEDLINE | ID: mdl-31231996

RESUMO

Cancer is one of the most important diseases threatening human health. Frequently-used traditional cancer treatment methods, like radiotherapy, chemotherapy and surgery, have serious toxic side effects and limitations. The widely-used drug delivery carriers (liposomes, nanoparticles, etc.) have also possessed many issues such as drug leakage and incomplete loading in the late clinical stage. Currently, using tumor-targeting vectors to deliver anti-tumor drugs or small molecules is one of the promising strategies for mediating safe and effective tumor therapy. In recent years, bacterial-derived non-replicating minicells, which are nanoscale non-nucleated cells produced during abnormal bacterial division, have got more and more attention. With a diameter of 200-400 nm, minicells have a large drug loading capacity. Meanwhile, the surface of minicells are able to be modified to load the assembly of antibodies/ligands that bind to tumor cell surface specific antigens or receptors, which can significantly improve tumor targeting of minicells. This tumor-targeting nanomaterials of minicells not only are used to deliver anti-tumor chemotherapeutic drugs, functional nucleic acids or plasmids encoding functional small molecules to mammalian cells, but also greatly increase drug loading and reduce drug penetration. Thus, the use of minicells combining with chemical therapy could help reduce the toxicity and maximize the effectiveness of the drug in the body. This paper summarizes the research and development of production purification, drug loading, tumor cells targeting, and internalization process of minicells, as well as its use in the delivery of anti-tumor drugs, to provide some information for the development and utilization of minicell carriers.


Assuntos
Nanopartículas , Neoplasias , Animais , Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Humanos , Plasmídeos
6.
Cancer Lett ; 448: 168-181, 2019 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-30753837

RESUMO

Bacteria have been investigated as anti-tumor therapeutic agents for more than a century, since Coley first observed successful curing of a patient with inoperable cancer by injection of streptococcal organisms. Previous studies have demonstrated that some obligate or facultative anaerobes can selectively accumulate and proliferate within tumors and suppress their growth. Developments in molecular biology as well as the complete genome sequencing of many bacterial species have increased the applicability of bacterial organisms for cancer treatment. In particular, the facultative anaerobe Salmonella Typhimurium has been widely studied and genetically engineered to improve its tumor-targeting ability as well as to reduce bacterial virulence. Moreover, the effectiveness of engineered attenuated S. Typhimurium strains employed as live delivery vectors of various anti-tumor therapeutic agents or combined with other therapies has been evaluated in a large number of animal experiments. The well-known S. Typhimurium mutant VNP20009 and its derivative strain TAPET-CD have even been applied in human clinical trials. However, Salmonella-mediated cancer therapies have not achieved the expected success, except in animal experiments. Many problems remain to be solved to exploit more promising strategies for combatting cancer with Salmonella bacteria. Here, we summarize the promising studies regarding cancer therapy mediated by Salmonella bacteria and highlight the main mechanisms of Salmonella anti-tumor activities.


Assuntos
Antineoplásicos/uso terapêutico , Vacinas Anticâncer/uso terapêutico , Neoplasias/terapia , Salmonella typhimurium/imunologia , Vacinas Atenuadas/uso terapêutico , Animais , Humanos
7.
Cancer Gene Ther ; 25(7-8): 167-183, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29755110

RESUMO

Salmonella typhimurium (hereafter S. typhimurium), as Gram-negative facultative anaerobic bacteria, are good candidates for cancer therapy and delivering therapeutic antitumor agents. However, it is necessary to reduce the virulence of such bacteria and enhance their tumor-targeting ability, and their immunostimulatory ability to induce tumor cell apoptosis. In this study, we constructed a S. typhimurium mutant named S634 harboring aroA mutation and additional mutations involved in modifications of lipid A. Upon intraperitoneal infection in mice, the aroA-deficient strain S634 showed greatly attenuated virulence and preferential accumulation within tumor tissue. We next investigated the ability of S636, the asd mutant derivative of S634, to deliver the anti-angiogenic agent "endostatin" (S636/pES) and to inhibit tumor growth in mouse CT26 colon carcinoma and B16F10 melanoma models. S636/pES-treated tumor-bearing mice showed suppressed tumor growth and prolonged survival, compared to mice treated with either the bacteria carrying empty plasmids or PBS intraperitoneally. Immunohistochemical studies demonstrated that, when tumor-bearing mice were infected with S636/pES, Salmonella colonization and endostatin expression were accompanied by the increase of apoptosis level and suppression of tumor angiogenesis within tumor tissues. Our findings showed that endostatin gene therapy delivered by attenuated S. typhimurium displays therapeutic antitumor effects in murine tumor models.


Assuntos
Neoplasias do Colo , Técnicas de Transferência de Genes , Terapia Genética/métodos , Melanoma , Neoplasias Experimentais , Salmonella typhimurium , Animais , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/terapia , Endostatinas , Melanoma/genética , Melanoma/metabolismo , Melanoma/terapia , Camundongos , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/terapia , Salmonella typhimurium/genética , Salmonella typhimurium/metabolismo
8.
Front Microbiol ; 7: 623, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27199951

RESUMO

Urinary tract infections (UTIs) are one of the most common bacterial infections in humans, causing cystitis, pyelonephritis, and renal failure. Uropathogenic Escherichia coli (UPEC) is the leading cause of UTIs. Accurate and rapid discrimination of UPEC lineages is useful for epidemiological surveillance. Fimbriae are necessary for the adherence of UPEC strains to host uroepithelia, and seem to be abundant and diverse in UPEC strains. By analyzing all the possible fimbrial operons in UPEC strains, we found that closely related strains had similar types of chaperone-usher fimbriae, and the diversity of fimbrial genes was higher than that of multilocus sequence typing (MLST) genes. A typing assay based on the polymorphism of four gene sequences (three fimbrial genes and one housekeeping gene) and the diversity of fimbriae present was developed. By comparison with the MLST, whole-genome sequence (WGS) and fumC/fimH typing methods, this was shown to be accurate and have high resolution, and it was also relatively inexpensive and easy to perform. The assay can supply more discriminatory information for UPEC lineages, and have the potential to be applied in epidemiological surveillance of UPEC isolates.

9.
Vet Microbiol ; 186: 189-98, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-27016773

RESUMO

DNA vaccines are widely used to prevent and treat infectious diseases, cancer and autoimmune diseases; however, their relatively low immunogenicity is an obstacle to their use. In this study, we constructed a novel and universal DNA vaccine vector (pSS898) that can be used to build DNA vaccines against duck enteritis virus (DEV) and other viruses that require DNA vaccines to provide protection. This vaccine vector has many advantages, including innate immunogenicity, efficient nuclear trafficking and resistance to attack from nucleases. UL24 and tgB from DEV were chosen as the antigens, and the heat labile enterotoxin B subunit (LTB) from Escherichia coli and the IL-2 gene (DuIL-2) from duck were used as adjuvants for the construction of DNA vaccine plasmids. Ducklings that were orally immunized with S739 (Salmonella Typhimurium Δasd-66 Δcrp-24 Δcya-25) and harboring these DEV DNA vaccines produced strong mucosal and systemic immune responses, and they resisted an otherwise lethal DEV challenge. More importantly, S739 (UL24-LTB) provided 90% protection after a priming-boost immunization. This study shows that our novel and universal DNA vaccine vector can be used efficiently in practical applications and may provide a promising method of orally inoculating ducks with a DEV DNA vaccine delivered by attenuated Salmonella Typhimurium for prevention of DVE.


Assuntos
Patos/imunologia , Infecções por Herpesviridae/veterinária , Doenças das Aves Domésticas/prevenção & controle , Salmonella typhimurium/imunologia , Vacinação/métodos , Vacinas de DNA , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Antígenos Virais/imunologia , Células COS , Chlorocebus aethiops , Patos/genética , Patos/virologia , Escherichia coli/genética , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/mortalidade , Infecções por Herpesviridae/prevenção & controle , Imunidade nas Mucosas/imunologia , Mardivirus/imunologia , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/mortalidade , Doenças das Aves Domésticas/virologia , Análise de Sobrevida
10.
Sheng Wu Gong Cheng Xue Bao ; 32(5): 565-576, 2016 May 25.
Artigo em Chinês | MEDLINE | ID: mdl-29019195

RESUMO

Tumor is a neoplasm formed by the abnormal proliferation of local tissue cells under the effects of different tumorigenic factors. Tumor-therapy has always been a difficult clinical issue, while regular cancer treatments, such as radiotherapy, chemotherapy and surgery, have obvious limitations. Earlier studies have shown that some obligate anaerobes or facultative anaerobes have anti-tumor effects, for example, Salmonella typhymurium as facultative anaerobic bacteria can selectively colonize tumors and inhibit their growth. Besides, Salmonella has many advantages in tumor-therapy. In the past decade or two, many researchers have carried out genetic manipulation to attenuate the virulence of Salmonella, to improve their specificity of tumor colonization and specially to use attenuated Salmonella as carriers to deliver a variety of anti-tumor therapeutic molecules, and these genetically modified Salmonella have shown good anti-tumor effects in many animal experiments. Along with further research of Salmonella-mediated antitumor treatment, applications of genetically modified Salmonella for more effective tumor-therapy are promising. We reviewed the anti-tumor mechanisms of Salmonella, the research progress in tumor-therapy using genetically modified Salmonella, and current problems and possible solutions.


Assuntos
Microrganismos Geneticamente Modificados , Neoplasias/terapia , Salmonella/genética , Animais , Humanos , Virulência
11.
Mar Drugs ; 13(6): 3388-406, 2015 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-26023843

RESUMO

Escherichia coli BL21 (DE3) and its derivatives are widely used for the production of recombinant proteins, but these purified proteins are always contaminated with lipopolysaccharide (LPS). LPS is recognized by the toll-like receptor 4 and myeloid differentiation factor 2 complex of mammalian immune cells and leads to release of pro-inflammatory cytokines. It is a vital step to remove LPS from the proteins before use for therapeutic purpose. In this study, we constructed BL21 (DE3) ∆msbB28 ∆pagP38 mutant, which produces a penta-acylated LPS with reduced endotoxicity. The plasmids harboring pagL and/or lpxE were then introduced into this mutant to further modify the LPS. The new strain (S004) carrying plasmid pQK004 (pagL and lpxE) produced mono-phosphoryated tetra-acylated lipid A, which induces markedly less production of tumor necrosis factor-α in the RAW264.7 and IL-12 in the THP1, but still retains ability to produce recombinant proteins. This study provides a strategy to decrease endotoxic activity of recombinant proteins purified from E. coli BL21 backgrounds and a feasible approach to modify lipid A structure for alternative purposes such as mono-phosphoryl lipid A (MPL) as vaccine adjuvants.


Assuntos
Endotoxinas/toxicidade , Escherichia coli/genética , Lipídeo A/química , Lipopolissacarídeos/química , Proteínas Recombinantes/isolamento & purificação , Animais , Linhagem Celular , Escherichia coli/metabolismo , Estudos de Viabilidade , Humanos , Mediadores da Inflamação/metabolismo , Interleucina-12/metabolismo , Lipopolissacarídeos/toxicidade , Camundongos , Mutação , Plasmídeos/genética , Proteínas Recombinantes/toxicidade , Fator de Necrose Tumoral alfa/metabolismo
12.
Int J Med Microbiol ; 304(3-4): 431-43, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24631214

RESUMO

Vesicle shedding from bacteria is a universal process in most Gram-negative bacteria and a few Gram-positive bacteria. In this report, we isolate extracellular membrane vesicles (MVs) from the supernatants of Gram-positive pathogen Clostridium perfringens (C. perfringens). We demonstrated vesicle production in a variety of virulent and nonvirulent type A strains. MVs did not contain alpha-toxin and NetB toxin demonstrated by negative reaction to specific antibody and absence of specific proteins identified by LC-MS/MS. C. perfringens MVs contained DNA components such as 16S ribosomal RNA gene (16S rRNA), alpha-toxin gene (plc) and the perfringolysin O gene (pfoA) demonstrated by PCR. We also identified a total of 431 proteins in vesicles by 1-D gel separation and LC-MS/MS analysis. In vitro studies demonstrated that vesicles could be internalized into murine macrophage RAW264.7 cells without direct cytotoxicity effects, causing release of inflammation cytokines including granulocyte colony stimulating factor (G-CSF), tumor necrosis factor-alpha (TNF-α) and interleukin-1 (IL-1), which could also be detected in mice injected with MVs through intraperitoneal (i.p.) route. Mice immunized with C. perfringens MVs produced high titer IgG, especially IgG1, antibodies against C. perfringens membrane proteins. However, this kind of antibody could not provide protection in mice following challenge, though it could slightly postpone the time of death. Our results indicate that release of MVs from C. perfringens could provide a previously unknown mechanism to induce release of inflammatory cytokines, especially TNF-α, these findings may contribute to a better understanding of the pathogenesis of C. perfringens infection.


Assuntos
Imunidade Adaptativa , Clostridium perfringens/imunologia , Imunidade Inata , Vesículas Secretórias/imunologia , Animais , Anticorpos Antibacterianos/sangue , Proteínas de Bactérias/análise , Linhagem Celular , Cromatografia Líquida , Citocinas/metabolismo , DNA Bacteriano/análise , DNA Ribossômico/análise , Eletroforese em Gel de Poliacrilamida , Endocitose , Feminino , Imunoglobulina G/sangue , Macrófagos/imunologia , Camundongos Endogâmicos BALB C , Vesículas Secretórias/química , Espectrometria de Massas em Tandem
13.
Pathog Dis ; 70(3): 390-400, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24532573

RESUMO

Clostridium perfringens is an important Gram-positive pathogen responsible for food poisoning, necrotic enteritis, gas gangrene, and even death. Escherichia coli Nissle 1917 (EcN) is a well-characterized probiotic strain with demonstrated benefits. In this study, we evaluated the effects of EcN on growth, toxin production, biofilm formation, and inflammatory cytokine responses of C. perfringens. In vitro co-culture experiments demonstrated that EcN inhibited growth, gas production, and toxin production (α-toxin and NetB) of C. perfringens in a dose-dependent manner. The growth inhibition effect was not observed when C. perfringens was incubated with EcN cell-free supernatants (CFSE), suggesting that growth inhibition was caused by nutrition competition during co-incubation. In vitro studies demonstrated that pre-incubation with EcN did not inhibit C. perfringens attachment to Caco-2 cells, but did reduce C. perfringens total number, toxin production, and cytotoxicity after 24 h. The similar growth inhibition results were also observed during the formation of C. perfringens biofilm. Finally, pre-incubation of EcN with RAW264.7 cells significantly decreased the production of inflammatory cytokines caused by the introduction of C. perfringens. Our results indicate that EcN can inhibit many of the pathological effects of C. perfringens in vitro conditions.


Assuntos
Biofilmes/crescimento & desenvolvimento , Infecções por Clostridium/metabolismo , Infecções por Clostridium/microbiologia , Clostridium perfringens/fisiologia , Citocinas/metabolismo , Escherichia coli/fisiologia , Animais , Antibiose , Linhagem Celular , Interações Hospedeiro-Patógeno , Humanos , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Interações Microbianas , Probióticos
14.
Infect Immun ; 79(12): 5027-38, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21930761

RESUMO

Lipopolysaccharide (LPS), composed of lipid A, core, and O-antigen, is a major virulence factor of Salmonella enterica serovar Typhimurium, with lipid A being a major stimulator to induce the proinflammatory response via the Toll-like receptor 4 (TLR4)-MD2-CD14 pathway. While Salmonella msbB mutants lacking the myristate chain in lipid A were investigated widely as an anticancer vaccine, inclusion of the msbB mutation in a Salmonella vaccine to deliver heterologous antigens has not yet been investigated. We introduced the msbB mutation alone or in combination with mutations in other lipid A acyl chain modification genes encoding PagL, PagP, and LpxR into wild-type S. enterica serovar Typhimurium. The msbB mutation reduced virulence, while the pagL, pagP, and lpxR mutations did not affect virulence in the msbB mutant background when administered orally to BALB/c mice. Also, all mutants exhibited sensitivity to polymyxin B but did not display sensitivity to deoxycholate. LPS derived from msbB mutants induced less inflammatory responses in human Mono Mac 6 and murine macrophage RAW264.7 cells in vitro. However, an msbB mutant did not decrease the induction of inflammatory responses in mice compared to the levels induced by the wild-type strain, whereas an msbB pagP mutant induced less inflammatory responses in vivo. The mutations were moved to an attenuated Salmonella vaccine strain to evaluate their effects on immunogenicity. Lipid A modification caused by the msbB mutation alone and in combination with pagL, pagP, and lpxR mutations led to higher IgA production in the vaginal tract but still retained the same IgG titer level in serum to PspA, a test antigen from Streptococcus pneumoniae, and to outer membrane proteins (OMPs) from Salmonella.


Assuntos
Proteínas de Bactérias/genética , Salmonelose Animal/prevenção & controle , Vacinas contra Salmonella/imunologia , Salmonella typhimurium/genética , Salmonella typhimurium/imunologia , Animais , Antibacterianos/farmacologia , Proteínas de Bactérias/metabolismo , Linhagem Celular , Humanos , Imunidade Inata , Lipoilação , Camundongos , Mutação , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/patogenicidade , Virulência
15.
J Immunol ; 187(1): 412-23, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21632711

RESUMO

The development of safe live, attenuated Salmonella vaccines may be facilitated by detoxification of its LPS. Recent characterization of the lipid A 1-phosphatase, LpxE, from Francisella tularensis allowed us to construct recombinant, plasmid-free strains of Salmonella that produce predominantly 1-dephosphorylated lipid A, similar to the adjuvant approved for human use. Complete lipid A 1-dephosphorylation was also confirmed under low pH, low Mg(2+) culture conditions, which induce lipid A modifications. LpxE expression in Salmonella reduced its virulence in mice by five orders of magnitude. Moreover, mice inoculated with these detoxified strains were protected against wild-type challenge. Candidate Salmonella vaccine strains synthesizing pneumococcal surface protein A (PspA) were also confirmed to possess nearly complete lipid A 1-dephosphorylation. After inoculation by the LpxE/PspA strains, mice produced robust levels of anti-PspA Abs and showed significantly improved survival against challenge with wild-type Streptococcus pneumoniae WU2 compared with vector-only-immunized mice, validating Salmonella synthesizing 1-dephosphorylated lipid A as an Ag-delivery system.


Assuntos
Adjuvantes Imunológicos/biossíntese , Endotoxinas/farmacologia , Lipídeo A/análogos & derivados , Lipopolissacarídeos/biossíntese , Proteínas de Membrana/genética , Monoéster Fosfórico Hidrolases/genética , Salmonella typhimurium/imunologia , Salmonella typhimurium/metabolismo , Adjuvantes Imunológicos/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Endotoxinas/imunologia , Feminino , Humanos , Lipídeo A/biossíntese , Lipídeo A/genética , Lipídeo A/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Monoéster Fosfórico Hidrolases/metabolismo , Fosforilação/imunologia , Coelhos , Vacinas contra Salmonella/genética , Vacinas contra Salmonella/imunologia , Vacinas contra Salmonella/metabolismo , Salmonella enterica/genética , Salmonella enterica/imunologia , Salmonella enterica/metabolismo , Salmonella typhimurium/genética , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/imunologia , Streptococcus pneumoniae/metabolismo , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA