Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.012
Filtrar
1.
J Med Chem ; 67(9): 7458-7469, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38634150

RESUMEN

Adjuvant is an integral part of all vaccine formulations but only a few adjuvants with limited efficacies or application scopes are available. Thus, developing more robust and diverse adjuvants is necessary. To this end, a new class of adjuvants having α- and ß-rhamnose (Rha) attached to the 1- and 6'-positions of monophosphoryl lipid A (MPLA) was designed, synthesized, and immunologically evaluated in mice. The results indicated a synergistic effect of MPLA and Rha, two immunostimulators that function via interacting with toll-like receptor 4 and recruiting endogenous anti-Rha antibodies, respectively. All the tested MPLA-Rha conjugates exhibited potent adjuvant activities to promote antibody production against both protein and carbohydrate antigens. Overall, MPLA-α-Rha exhibited better activities than MPLA-ß-Rha, and 6'-linked conjugates were slightly better than 1-linked ones. Particularly, MPLA-1-α-Rha and MPLA-6'-α-Rha were the most effective adjuvants in promoting IgG antibody responses against protein antigen keyhole limpet hemocyanin and carbohydrate antigen sTn, respectively.


Asunto(s)
Lípido A , Ramnosa , Lípido A/análogos & derivados , Lípido A/química , Lípido A/farmacología , Lípido A/inmunología , Animales , Ramnosa/química , Ramnosa/inmunología , Ramnosa/farmacología , Ratones , Adyuvantes de Vacunas/química , Adyuvantes de Vacunas/farmacología , Femenino , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/síntesis química , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina G/sangre , Ratones Endogámicos BALB C , Hemocianinas/química , Hemocianinas/inmunología
2.
Vet Immunol Immunopathol ; 271: 110743, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38522410

RESUMEN

Equine influenza is a contagious respiratory disease caused by H3N8 type A influenza virus. Vaccination against equine influenza is conducted regularly; however, infection still occurs globally because of the short immunity duration and suboptimal efficacy of current vaccines. Hence the objective of this study was to investigate whether an adjuvant combination can improve immune responses to equine influenza virus (EIV) vaccines. Seventy-two mice were immunized with an EIV vaccine only or with monophosphoryl lipid A (MPL), polyinosinic-polycytidylic acid (Poly I:C), or MPL + Poly I:C. Prime immunization was followed by boost immunization after 2 weeks. Mice were euthanized at 4, 8, and 32 weeks post-prime immunization, respectively. Sera were collected to determine humoral response. Bone marrow, spleen, and lung samples were harvested to determine memory cell responses, antigen-specific T-cell proliferation, and lung viral titers. MPL + Poly I:C resulted in the highest IgG, IgG1, and IgG2a antibodies and hemagglutination inhibition titers among the groups and sustained their levels until 32 weeks post-prime immunization. The combination enhanced memory B cell responses in the bone marrow and spleen. At 8 weeks post-prime immunization, the combination induced higher CD8+ central memory T cell frequencies in the lungs and CD8+ central memory T cells in the spleen. In addition, the combination group exhibited enhanced antigen-specific T cell proliferation, except for CD4+ T cells in the lungs. Our results demonstrated improved immune responses when using MPL + Poly I:C in EIV vaccines by inducing enhanced humoral responses, memory cell responses, and antigen-specific T cell proliferation.


Asunto(s)
Adyuvantes Inmunológicos , Subtipo H3N8 del Virus de la Influenza A , Vacunas contra la Influenza , Lípido A , Lípido A/análogos & derivados , Infecciones por Orthomyxoviridae , Poli I-C , Animales , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación , Poli I-C/farmacología , Poli I-C/administración & dosificación , Lípido A/farmacología , Lípido A/administración & dosificación , Lípido A/inmunología , Ratones , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/veterinaria , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/farmacología , Femenino , Subtipo H3N8 del Virus de la Influenza A/inmunología , Anticuerpos Antivirales/sangre , Caballos/inmunología , Enfermedades de los Caballos/inmunología , Enfermedades de los Caballos/prevención & control , Enfermedades de los Caballos/virología , Inmunoglobulina G/sangre , Memoria Inmunológica
3.
Int Immunol ; 36(1): 33-43, 2024 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-38006376

RESUMEN

We previously demonstrated that Alcaligenes-derived lipid A (ALA), which is produced from an intestinal lymphoid tissue-resident commensal bacterium, is an effective adjuvant for inducing antigen-specific immune responses. To understand the immunologic characteristics of ALA as a vaccine adjuvant, we here compared the adjuvant activity of ALA with that of a licensed adjuvant (monophosphoryl lipid A, MPLA) in mice. Although the adjuvant activity of ALA was only slightly greater than that of MPLA for subcutaneous immunization, ALA induced significantly greater IgA antibody production than did MPLA during nasal immunization. Regarding the underlying mechanism, ALA increased and activated CD11b+ CD103- CD11c+ dendritic cells in the nasal tissue by stimulating chemokine responses. These findings revealed the superiority of ALA as a mucosal adjuvant due to the unique immunologic functions of ALA in nasal tissue.


Asunto(s)
Alcaligenes , Lípido A , Animales , Ratones , Lípido A/farmacología , Adyuvantes Inmunológicos/farmacología , Células Dendríticas
4.
J Glob Antimicrob Resist ; 33: 101-108, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36906175

RESUMEN

OBJECTIVES: Colistin-resistant Gram-negative pathogens have become a serious worldwide medical problem. This study was designed to reveal the effects of an intrinsic phosphoethanolamine transferase from Acinetobacter modestus on Enterobacterales. METHODS: A strain of colistin-resistant A. modestus was isolated from a sample of nasal secretions taken in 2019 from a hospitalised pet cat in Japan. The whole genome was sequenced by next generation sequencing, and transformants of Escherichia coli, Klebsiella pneumoniae, and Enterobacter cloacae harbouring the phosphoethanolamine transferase-encoding gene from A. modestus were constructed. Lipid A modification in E. coli transformants was analysed using electrospray ionization mass spectrometry. RESULTS: Sequencing of the entire genome revealed that the isolate harboured a phosphoethanolamine transferase-encoding gene, eptA_AM, on its chromosome. Transformants of E. coli, K. pneumoniae, and E. cloacae harbouring both the promoter and eptA_AM gene from A. modestus had 32-fold, 8-fold, and 4-fold higher minimum inhibitory concentrations (MICs) for colistin, respectively, than transformants harbouring a control vector. The genetic environment surrounding eptA_AM in A. modestus was similar to that surrounding eptA_AM in Acinetobacter junii and Acinetobacter venetianus. Electrospray ionization mass spectrometry analysis revealed that EptA_AM modified lipid A in Enterobacterales. CONCLUSION: This is the first report to describe the isolation of an A. modestus strain in Japan and show that its intrinsic phosphoethanolamine transferase, EptA_AM, contributes to colistin resistance in Enterobacterales and A. modestus.


Asunto(s)
Colistina , Escherichia coli , Animales , Gatos , Colistina/farmacología , Escherichia coli/genética , Lípido A/farmacología , Etanolaminofosfotransferasa/genética , Proteínas Bacterianas/genética , Farmacorresistencia Bacteriana/genética , Antibacterianos/farmacología , Klebsiella pneumoniae
5.
Int Immunopharmacol ; 117: 109852, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36806039

RESUMEN

Alcaligenes faecalis was previously identified as an intestinal lymphoid tissue-resident commensal bacteria, and our subsequent studies showed that lipopolysaccharide and its core active element (i.e., lipid A) have a potent adjuvant activity to promote preferentially antigen-specific Th17 response and antibody production. Here, we compared A. faecalis lipid A (ALA) with monophosphoryl lipid A, a licensed lipid A-based adjuvant, to elucidate the immunological mechanism underlying the adjuvant properties of ALA. Compared with monophosphoryl lipid A, ALA induced higher levels of MHC class II molecules and costimulatory CD40, CD80, and CD86 on dendritic cells (DCs), which in turn resulted in strong T cell activation. Moreover, ALA more effectively promoted the production of IL-6 and IL-23 from DCs than did monophosphoryl lipid A, thus leading to preferential induction of Th17 and Th1 cells. As underlying mechanisms, we found that the ALA-TLR4 axis stimulated both MyD88- and TRIF-mediated signaling pathways, whereas monophosphoryl lipid A was biased toward TRIF signaling. These findings revealed the effects of ALA on DCs and T cells and its induction pattern on signaling pathways.


Asunto(s)
Lípido A , Factor 88 de Diferenciación Mieloide , Lípido A/farmacología , Lípido A/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Receptor Toll-Like 4/metabolismo , Presentación de Antígeno , Alcaligenes/metabolismo , Transducción de Señal , Adyuvantes Inmunológicos/farmacología , Diferenciación Celular , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Células Dendríticas
6.
Nat Commun ; 14(1): 140, 2023 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-36627272

RESUMEN

Species within the Enterobacter cloacae complex (ECC) include globally important nosocomial pathogens. A three-year study of ECC in Germany identified Enterobacter xiangfangensis as the most common species (65.5%) detected, a result replicated by examining a global pool of 3246 isolates. Antibiotic resistance profiling revealed widespread resistance and heteroresistance to the antibiotic colistin and detected the mobile colistin resistance (mcr)-9 gene in 19.2% of all isolates. We show that resistance and heteroresistance properties depend on the chromosomal arnBCADTEF gene cassette whose products catalyze transfer of L-Ara4N to lipid A. Using comparative genomics, mutational analysis, and quantitative lipid A profiling we demonstrate that intrinsic lipid A modification levels are genospecies-dependent and governed by allelic variations in phoPQ and mgrB, that encode a two-component sensor-activator system and specific inhibitor peptide. By generating phoPQ chimeras and combining them with mgrB alleles, we show that interactions at the pH-sensing interface of the sensory histidine kinase phoQ dictate arnBCADTEF expression levels. To minimize therapeutic failures, we developed an assay that accurately detects colistin resistance levels for any ECC isolate.


Asunto(s)
Colistina , Lípido A , Colistina/farmacología , Colistina/uso terapéutico , Lípido A/química , Lípido A/farmacología , Proteínas Bacterianas/metabolismo , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Enterobacter/genética , Farmacorresistencia Bacteriana/genética , Pruebas de Sensibilidad Microbiana
7.
Methods Mol Biol ; 2613: 33-53, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36587069

RESUMEN

Lipopolysaccharide (LPS), a cell surface component of Gram-negative bacteria, and its active principle, lipid A, have immunostimulatory properties and thus potential to act as adjuvants. However, canonical LPS acts as an endotoxin by hyperstimulating the immune response. Therefore, it is necessary to structurally modify LPS and lipid A to minimize toxicity while maintaining adjuvant effects for use as vaccine adjuvants. Various studies have focused on the chemical synthetic method of lipid As and their structure-activity relationship, which are reviewed in this chapter.


Asunto(s)
Lípido A , Lipopolisacáridos , Lipopolisacáridos/farmacología , Lípido A/farmacología , Lípido A/química , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/química , Bacterias/metabolismo , Inmunidad
8.
Atherosclerosis ; 358: 1-11, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36049289

RESUMEN

BACKGROUND AND AIMS: Gut microbial lipopolysaccharide (LPS) induces endotoxemia, an independent risk factor for cardiovascular disease (CVD). However, no studies have demonstrated how structural differences in each bacterial LPS contribute to endotoxemia. Here, we investigated the effects of different acyl chains in the lipid A moiety of LPS on endotoxemia and the subsequent immune response and atherosclerotic plaque formation. METHODS: Apoe-/- mice were intraperitoneally administered 2 mg/kg of Escherichia coli-derived LPS (E. LPS, as a representative of hexa-acylated lipid A), Bacteroides-derived LPS (B. LPS, as a representative of penta- or tetra-acylated lipid A), or saline (control) once a week, six times. An immunohistological assessment was performed on plaque sections. RESULTS: E. LPS administration induced endotoxemia, but B. LPS and saline did not. In E. LPS-treated mice, total plaque areas in the aortic root were significantly increased, and neutrophil accumulation and increased formation of neutrophil extracellular traps (NETs) were observed at the plaque lesions, but not in B. LPS-treated mice. A single dose of E. LPS significantly increased the accumulation of neutrophils in plaque lesions on day 3, and NET formation on day 7. E. LPS also increased interleukin-1 beta (IL-1ß) production in plaque lesions on day 7. Furthermore, NET formation and IL-1ß production were also observed in human coronary plaques. CONCLUSIONS: We identified a previously unknown link between structural differences in LPS and atherosclerosis. Lowering microbial LPS activity may reduce NET formation in plaques and prevent CVD progression.


Asunto(s)
Aterosclerosis , Endotoxemia , Placa Aterosclerótica , Animales , Apolipoproteínas E , Aterosclerosis/patología , Endotoxemia/inducido químicamente , Humanos , Interleucina-1beta/farmacología , Lípido A/farmacología , Lípido A/uso terapéutico , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Neutrófilos , Placa Aterosclerótica/patología
9.
Biomed Pharmacother ; 155: 113682, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36095964

RESUMEN

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.


Asunto(s)
Neoplasias , Salmonella typhimurium , Ratones , Animales , Antígenos O/metabolismo , Lipopolisacáridos/farmacología , Endostatinas/farmacología , Lípido A/metabolismo , Lípido A/farmacología , Integrina alfaV/metabolismo , Péptidos/farmacología , Péptidos/metabolismo
10.
J Neuroinflammation ; 19(1): 12, 2022 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-34996472

RESUMEN

BACKGROUND: Innate immune pre-stimulation can prevent the development of depression-like behaviors in chronically stressed mice; however, whether the same stimulation prevents the development of anxiety-like behaviors in animals remains unclear. We addressed this issue using monophosphoryl lipid A (MPL), a derivative of lipopolysaccharide (LPS) that lacks undesirable properties of LPS but still keeps immune-enhancing activities. METHODS: The experimental mice were pre-injected intraperitoneally with MPL before stress exposure. Depression was induced through chronic social defeat stress (CSDS). Behavioral tests were conducted to identify anxiety-like behaviors. Real-time polymerase chain reaction (PCR) and biochemical assays were employed to examine the gene and protein expression levels of pro-inflammatory markers. RESULTS: A single MPL injection at the dose of 400 and 800 µg/kg 1 day before stress exposure prevented CSDS-induced anxiety-like behaviors, and a single MPL injection (400 µg/kg) five but not 10 days before stress exposure produced similar effect. The preventive effect of MPL on anxiety-like behaviors was also observed in CSDS mice who received a second MPL injection 10 days after the first MPL injection or a 4 × MPL injection 10 days before stress exposure. MPL pre-injection also prevented the production of pro-inflammatory cytokines in the hippocampus and medial prefrontal cortex in CSDS mice, and inhibiting the central immune response by minocycline pretreatment abrogated the preventive effect of MPL on CSDS-induced anxiety-like behaviors and pro-inflammatory cytokine productions in the brain. CONCLUSIONS: Pre-stimulation of the innate immune system by MPL can prevent chronic stress-induced anxiety-like behaviors and neuroinflammatory responses in the brain in mice.


Asunto(s)
Ansiedad/inmunología , Inmunidad Innata/efectos de los fármacos , Lípido A/análogos & derivados , Corteza Prefrontal/efectos de los fármacos , Derrota Social , Estrés Psicológico/inmunología , Animales , Depresión/inmunología , Lípido A/farmacología , Lipopolisacáridos/farmacología , Masculino , Ratones , Corteza Prefrontal/inmunología , Conducta Social
11.
PLoS One ; 16(12): e0259301, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34855754

RESUMEN

Systemic interleukin-12 (IL12) anti-tumor therapy is highly potent but has had limited utility in the clinic due to severe toxicity. Here, we present two IL12-expressing vector platforms, both of which can overcome the deficiencies of previous systemic IL12 therapies: 1) an integrating lentiviral vector, and 2) a self-replicating messenger RNA formulated with polyethyleneimine. Intratumoral administration of either IL12 vector platform resulted in recruitment of immune cells, including effector T cells and dendritic cells, and the complete remission of established tumors in multiple murine models. Furthermore, concurrent intratumoral administration of the synthetic TLR4 agonist glucopyranosyl lipid A formulated in a stable emulsion (GLA-SE) induced systemic memory T cell responses that mediated complete protection against tumor rechallenge in all survivor mice (8/8 rechallenged mice), whereas only 2/6 total rechallenged mice treated with intratrumoral IL12 monotherapy rejected the rechallenge. Taken together, expression of vectorized IL12 in combination with a TLR4 agonist represents a varied approach to broaden the applicability of intratumoral immune therapies of solid tumors.


Asunto(s)
Glucósidos/farmacología , Memoria Inmunológica/efectos de los fármacos , Interleucina-12/genética , Lípido A/farmacología , Neoplasias Experimentales/inmunología , Receptor Toll-Like 4/agonistas , Animales , Linfocitos T CD8-positivos/inmunología , Femenino , Regulación de la Expresión Génica , Vectores Genéticos/administración & dosificación , Vectores Genéticos/farmacología , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/genética , Memoria Inmunológica/genética , Inmunoterapia/métodos , Interferón gamma/sangre , Interleucina-12/sangre , Interleucina-12/inmunología , Lentivirus/genética , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología
12.
Front Immunol ; 12: 701445, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34650551

RESUMEN

As viruses continue to mutate the need for rapid high titer neutralizing antibody responses has been highlighted. To meet these emerging threats, agents that enhance vaccine adjuvant activity are needed that are safe with minimal local or systemic side effects. To respond to this demand, we sought small molecules that would sustain and improve the protective effect of a currently approved adjuvant, monophosphoryl lipid A (MPLA), a Toll-like receptor 4 (TLR4) agonist. A lead molecule from a high-throughput screen, (N-(4-(2,5-dimethylphenyl)thiazol-2-yl)-4-(piperidin-1-ylsulfonyl)benzamide, was identified as a hit compound that sustained NF-κB activation by a TLR4 ligand, lipopolysaccharide (LPS), after an extended incubation (16 h). In vitro, the resynthesized compound (2D216) enhanced TLR4 ligand-induced innate immune activation and antigen presenting function in primary murine bone marrow-derived dendritic cells without direct activation of T cells. In vivo murine vaccination studies demonstrated that compound 2D216 acted as a potent co-adjuvant when used in combination with MPLA that enhanced antigen-specific IgG equivalent to that of AS01B. The combination adjuvant MPLA/2D216 produced Th1 dominant immune responses and importantly protected mice from lethal influenza virus challenge. 2D216 alone or 2D216/MPLA demonstrated minimal local reactogenicity and no systemic inflammatory response. In summary, 2D216 augmented the beneficial protective immune responses of MPLA as a co-adjuvant and showed an excellent safety profile.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/farmacología , Lípido A/análogos & derivados , Animales , Femenino , Virus de la Influenza A , Lípido A/inmunología , Lípido A/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae
13.
Molecules ; 26(20)2021 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-34684874

RESUMEN

Gram-negative bacterial cell surface component lipopolysaccharide (LPS) and its active principle, lipid A, exhibit immunostimulatory effects and have the potential to act as adjuvants. However, canonical LPS acts as an endotoxin by hyperstimulating the immune response. Therefore, LPS and lipid A must be structurally modified to minimize their toxic effects while maintaining their adjuvant effect for application as vaccine adjuvants. In the field of chemical ecology research, various biological phenomena occurring among organisms are considered molecular interactions. Recently, the hypothesis has been proposed that LPS and lipid A mediate bacterial-host chemical ecology to regulate various host biological phenomena, mainly immunity. Parasitic and symbiotic bacteria inhabiting the host are predicted to possess low-toxicity immunomodulators due to the chemical structural changes of their LPS caused by co-evolution with the host. Studies on the chemical synthesis and functional evaluation of their lipid As have been developed to test this hypothesis and to apply them to low-toxicity and safe adjuvants.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Bacterias/inmunología , Endotoxinas/inmunología , Lípido A/farmacología , Lipopolisacáridos/inmunología , Adyuvantes Inmunológicos/química , Animales , Bacterias/efectos de los fármacos , Endotoxinas/metabolismo , Humanos , Lípido A/química , Lipopolisacáridos/química
14.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34561306

RESUMEN

The COVID-19 pandemic highlights the importance of efficient and safe vaccine development. Vaccine adjuvants are essential to boost and tailor the immune response to the corresponding pathogen. To allow for an educated selection, we assessed the effect of different adjuvants on human monocyte-derived dendritic cells (DCs) and their ability to polarize innate and adaptive immune responses. In contrast to commonly used adjuvants, such as aluminum hydroxide, Toll-like receptor (TLR) agonists induced robust phenotypic and functional DC maturation. In a DC-lymphocyte coculture system, we investigated the ensuing immune reactions. While monophosphoryl lipid A synthetic, a TLR4 ligand, induced checkpoint inhibitors indicative for immune exhaustion, the TLR7/8 agonist Resiquimod (R848) induced prominent type-1 interferon and interleukin 6 responses and robust CTL, B-cell, and NK-cell proliferation, which is particularly suited for antiviral immune responses. The recently licensed COVID-19 vaccines, BNT162b and mRNA-1273, are both based on single-stranded RNA. Indeed, we could confirm that the cytokine profile induced by lipid-complexed RNA was almost identical to the pattern induced by R848. Although this awaits further investigation, our results suggest that their efficacy involves the highly efficient antiviral response pattern stimulated by the RNAs' TLR7/8 activation.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , COVID-19/inmunología , Células Dendríticas/inmunología , Inmunidad Celular/efectos de los fármacos , SARS-CoV-2/inmunología , Linfocitos T/inmunología , Adolescente , Adulto , Anciano , Femenino , Humanos , Imidazoles/farmacología , Lípido A/análogos & derivados , Lípido A/farmacología , Masculino , Persona de Mediana Edad , Receptores Toll-Like/inmunología
15.
Immunobiology ; 226(5): 152132, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34478947

RESUMEN

BACKGROUND: The monophosphoryl lipid A (MPLA) is a detoxified LPS derivative and an emerging safe immune adjuvant in human vaccine development. The adjuvant MPLA promotes antigen-presenting cell (APC) function and preferentially induces a Th1 response following vaccination. However, the mechanism by which the MPLA detoxicates and exerts its adjuvants effect on T-cell, particualrly the Th1 response is unknown. AIMS: We assessed the direct effects of MPLA on murine and human CD4+ T-cell proliferation and the profile of cytokine production ex vivo. RESULTS: We report that CD4+ T-cells only express functional TLR2 and TLR4 when activated by TCR stimulation, in particularly in the presence of IFNα. The activated T cells thereafter can respond directly to MPLA. MPLA does not affect T-cell proliferation in human T cells, but can induce a balanced Th1 cytokine profile in CD4+ T-cells by reducing the production of Th1 cytokines and enhancing the production of the regulatory cytokine IL-10. The MPLA-mediated regulatory effect on activated CD4+ T-cells is TLR2 and TLR4 dependent and can be abolished by the lipid A blocker polymyxin B. CONCLUSION: These data provide evidence, at least in part, for the safe induction of an appropriate level of Th1 response by adjuvant MPLA in human vaccine development.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Citocinas/inmunología , Lípido A/análogos & derivados , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 4/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Femenino , Humanos , Lípido A/farmacología , Ratones Endogámicos C3H , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética
16.
Front Immunol ; 12: 631797, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33815382

RESUMEN

Pro-inflammatory signaling mediated by Toll-like receptor 4 (TLR4)/myeloid differentiation-2 (MD-2) complex plays a crucial role in the instantaneous protection against infectious challenge and largely contributes to recovery from Gram-negative infection. Activation of TLR4 also boosts the adaptive immunity which is implemented in the development of vaccine adjuvants by application of minimally toxic TLR4 activating ligands. The modulation of pro-inflammatory responses via the TLR4 signaling pathway was found beneficial for management of acute and chronic inflammatory disorders including asthma, allergy, arthritis, Alzheimer disease pathology, sepsis, and cancer. The TLR4/MD-2 complex can recognize the terminal motif of Gram-negative bacterial lipopolysaccharide (LPS)-a glycophospholipid lipid A. Although immense progress in understanding the molecular basis of LPS-induced TLR4-mediated signaling has been achieved, gradual, and predictable TLR4 activation by structurally defined ligands has not yet been attained. We report on controllable modulation of cellular pro-inflammatory responses by application of novel synthetic glycolipids-disaccharide-based lipid A mimetics (DLAMs) having picomolar affinity for TLR4/MD-2. Using crystal structure inspired design we have developed endotoxin mimetics where the inherently flexible ß(1 → 6)-linked diglucosamine backbone of lipid A is replaced by a conformationally restricted α,α-(1↔1)-linked disaccharide scaffold. The tertiary structure of the disaccharide skeleton of DLAMs mirrors the 3-dimensional shape of TLR4/MD-2 bound E. coli lipid A. Due to exceptional conformational rigidity of the sugar scaffold, the specific 3D organization of DLAM must be preserved upon interaction with proteins. These structural factors along with specific acylation and phosphorylation pattern can ensure picomolar affinity for TLR4 and permit efficient dimerization of TLR4/MD-2/DLAM complexes. Since the binding pose of lipid A in the binding pocket of MD-2 (±180°) is crucial for the expression of biological activity, the chemical structure of DLAMs was designed to permit a predefined binding orientation in the binding groove of MD-2, which ensured tailored and species-independent (human and mice) TLR4 activation. Manipulating phosphorylation and acylation pattern at the sugar moiety facing the secondary dimerization interface allowed for adjustable modulation of the TLR4-mediated signaling. Tailored modulation of cellular pro-inflammatory responses by distinct modifications of the molecular structure of DLAMs was attained in primary human and mouse immune cells, lung epithelial cells and TLR4 transfected HEK293 cells.


Asunto(s)
Materiales Biomiméticos/farmacología , Disacáridos/farmacología , Inmunomodulación , Lípido A/farmacología , Animales , Materiales Biomiméticos/síntesis química , Materiales Biomiméticos/química , Citocinas/inmunología , Disacáridos/química , Escherichia coli , Células HEK293 , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Lípido A/química , Lipopolisacáridos/química , Lipopolisacáridos/farmacología , Antígeno 96 de los Linfocitos/química , Antígeno 96 de los Linfocitos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , FN-kappa B/inmunología , Transducción de Señal/efectos de los fármacos , Células THP-1 , Receptor Toll-Like 4/química , Receptor Toll-Like 4/inmunología
17.
Angew Chem Int Ed Engl ; 60(18): 10023-10031, 2021 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-33522128

RESUMEN

Alcaligenes faecalis is the predominant Gram-negative bacterium inhabiting gut-associated lymphoid tissues, Peyer's patches. We previously reported that an A. faecalis lipopolysaccharide (LPS) acted as a weak agonist for Toll-like receptor 4 (TLR4)/myeloid differentiation factor-2 (MD-2) receptor as well as a potent inducer of IgA without excessive inflammation, thus suggesting that A. faecalis LPS might be used as a safe adjuvant. In this study, we characterized the structure of both the lipooligosaccharide (LOS) and LPS from A. faecalis. We synthesized three lipid A molecules with different degrees of acylation by an efficient route involving the simultaneous introduction of 1- and 4'-phosphates. Hexaacylated A. faecalis lipid A showed moderate agonistic activity towards TLR4-mediated signaling and the ability to elicit a discrete interleukin-6 release in human cell lines and mice. It was thus found to be the active principle of the LOS/LPS and a promising vaccine adjuvant candidate.


Asunto(s)
Alcaligenes faecalis/química , Lípido A/química , Lipopolisacáridos/química , Animales , Conformación de Carbohidratos , Línea Celular , Humanos , Interleucina-6/antagonistas & inhibidores , Interleucina-6/metabolismo , Lípido A/farmacología , Lipopolisacáridos/aislamiento & purificación , Lipopolisacáridos/farmacología , Ratones , Receptor Toll-Like 4/agonistas
18.
J Surg Res ; 261: 74-84, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33421796

RESUMEN

BACKGROUND: Although the ability of ß-D-glucan and monophosphoryl lipid A (MPLA) to modulate immune responses has been studied in human primary cells, their effect on sterile inflammation models such as necrotizing pancreatitis has never been investigated. MATERIALS AND METHODS: 85 male New Zealand rabbits were assigned into following groups: A: control, B: pretreatment with ß-D-glucan 3 d before pancreatitis, C: pretreatment with MPLA 3 d before pancreatitis, D: pretreatment with ß-D-glucan and laminarin 3 d before pancreatitis, E: treatment with ß-D-glucan 1 d after pancreatitis, and F: MPLA 1 d after pancreatitis. Pancreatitis was induced by sodium taurocholate injection into the pancreatic duct and parenchyma. Survival was recorded for 21 d. On days 1, 3, and 7, blood was collected for amylase measurement. Peripheral blood mononuclear cells were isolated and stimulated for tumor necrosis factor alpha and interleukin 10 production. Pancreatic necrosis and tissue bacterial load were assessed. RESULTS: 21-d survival was prolonged after pretreatment or treatment with ß-D-glucan; this benefit was lost with laminarin administration. At sacrifice, pancreatic inflammatory alterations were more prominent in the control group. Bacterial load was lower after pretreatment or treatment with ß-D-glucan and MPLA. Tumor necrosis factor alpha production from stimulated peripheral blood mononuclear cells was significantly decreased, whereas interleukin 10 production remained unaltered after pretreatment or treatment with ß-D- glucan. CONCLUSIONS: ß-D-glucan reduces mortality of experimental pancreatitis in vivo. This is mediated through attenuation of cytokine production and prevention of bacterial translocation.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Inmunomodulación , Lípido A/análogos & derivados , Pancreatitis Aguda Necrotizante/tratamiento farmacológico , Proteoglicanos/uso terapéutico , Adyuvantes Inmunológicos/farmacología , Amilasas/sangre , Animales , Traslocación Bacteriana/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Glucanos , Lípido A/farmacología , Lípido A/uso terapéutico , Masculino , Pancreatitis Aguda Necrotizante/sangre , Pancreatitis Aguda Necrotizante/mortalidad , Proteoglicanos/farmacología , Conejos , Ácido Taurocólico , Factor de Necrosis Tumoral alfa/metabolismo
19.
Front Immunol ; 12: 748303, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35140704

RESUMEN

The Toll-like receptor 4 (TLR4)/myeloid differentiation protein-2 (MD-2) complex is considered the major receptor of the innate immune system to recognize lipopolysaccharides (LPSs). However, some atypical LPSs with different lipid A and core saccharide moiety structures and compositions than the well-studied enterobacterial LPSs can induce a TLR2-dependent response in innate immune cells. Ochrobactrum intermedium, an opportunistic pathogen, presents an atypical LPS. In this study, we found that O. intermedium LPS exhibits a weak inflammatory activity compared to Escherichia coli LPS and, more importantly, is a specific TLR4/TLR2 agonist, able to signal through both receptors. Molecular docking analysis of O. intermedium LPS predicts a favorable formation of a TLR2/TLR4/MD-2 heterodimer complex, which was experimentally confirmed by fluorescence resonance energy transfer (FRET) in cells. Interestingly, the core saccharide plays an important role in this interaction. This study reveals for the first time TLR4/TLR2 heterodimerization that is induced by atypical LPS and may help to escape from recognition by the innate immune system.


Asunto(s)
Endotoxinas/farmacología , Lipopolisacáridos/farmacología , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Línea Celular , Células HEK293 , Humanos , Inmunidad Innata/efectos de los fármacos , Inflamación/metabolismo , Lípido A/farmacología , Ratones Endogámicos C57BL , Ratones Noqueados , Simulación del Acoplamiento Molecular
20.
Cancer Immunol Immunother ; 70(3): 689-700, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32902663

RESUMEN

Monophosphoryl lipid A (MPLA) is a toll-like receptor 4 ligand that promotes immune activation in mice and humans, without undesired inflammation. Immunotherapy by the combining immune checkpoint blockade and MPLA has shown promising anti-cancer effects in both mice and humans. In this study, we explored how MPLA enhanced the anti-cancer effects of anti-PD-L1 antibodies (Abs). Anti-cancer immunity induced by the combination of anti-PD-L1 Abs and MPLA failed in CD4 and CD8 cell-depleted mice. Moreover, the combination treatment of anti-PD-L1 Abs and MPLA synergistically enhanced the activation of plasmacytoid dendritic cells (pDCs) in the mouse in vivo, while conventional DCs were not. In addition, mice treated with anti-PD-L1 Abs and MPLA were not protected from B16 melanoma by blockade of interferon-alpha receptor (IFNAR). The combination of anti-PD-L1 Abs and MPLA also promoted human peripheral blood pDC activation and induced IFN-α-dependent T cell activation. Therefore, these results demonstrate that MPLA enhances anti-PD-L1 Ab-mediated anti-cancer immunity through the activation and IFN-α production of pDCs.


Asunto(s)
Antígeno B7-H1/antagonistas & inhibidores , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Inhibidores de Puntos de Control Inmunológico/farmacología , Lípido A/análogos & derivados , Animales , Biomarcadores , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Citocinas/metabolismo , Células Dendríticas/metabolismo , Femenino , Humanos , Inmunofenotipificación , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Lípido A/farmacología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Melanoma Experimental , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...