Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
ACS Appl Bio Mater ; 7(6): 3877-3889, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38832760

ABSTRACT

Adjuvants and immunomodulators that effectively drive a Th17-skewed immune response are not part of the standard vaccine toolkit. Vaccine adjuvants and delivery technologies that can induce Th17 or Th1/17 immunity and protection against bacterial pathogens, such as tuberculosis (TB), are urgently needed. Th17-polarized immune response can be induced using agonists that bind and activate C-type lectin receptors (CLRs) such as macrophage inducible C-type lectin (Mincle). A simple but effective strategy was developed for codelivering Mincle agonists with the recombinant Mycobacterium tuberculosis fusion antigen, M72, using tunable silica nanoparticles (SNP). Anionic bare SNP, hydrophobic phenyl-functionalized SNP (P-SNP), and cationic amine-functionalized SNP (A-SNP) of different sizes were coated with three synthetic Mincle agonists, UM-1024, UM-1052, and UM-1098, and evaluated for adjuvant activity in vitro and in vivo. The antigen and adjuvant were coadsorbed onto SNP via electrostatic and hydrophobic interactions, facilitating multivalent display and delivery to antigen presenting cells. The cationic A-SNP showed the highest coloading efficiency for the antigen and adjuvant. In addition, the UM-1098-adsorbed A-SNP formulation demonstrated slow-release kinetics in vitro, excellent stability over 12 months of storage, and strong IL-6 induction from human peripheral blood mononuclear cells. Co-adsorption of UM-1098 and M72 on A-SNP significantly improved antigen-specific humoral and Th17-polarized immune responses in vivo in BALB/c mice relative to the controls. Taken together, A-SNP is a promising platform for codelivery and proper presentation of adjuvants and antigens and provides the basis for their further development as a vaccine delivery platform for immunization against TB or other diseases for which Th17 immunity contributes to protection.


Subject(s)
Antigens, Bacterial , Lectins, C-Type , Nanoparticles , Silicon Dioxide , Th17 Cells , Lectins, C-Type/metabolism , Lectins, C-Type/immunology , Lectins, C-Type/agonists , Nanoparticles/chemistry , Th17 Cells/immunology , Animals , Silicon Dioxide/chemistry , Mice , Antigens, Bacterial/immunology , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/chemistry , Mycobacterium tuberculosis/immunology , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/administration & dosage , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Particle Size , Materials Testing , Humans , Female , Membrane Proteins/immunology , Membrane Proteins/agonists
2.
Tetrahedron ; 1322023 Feb 13.
Article in English | MEDLINE | ID: mdl-36874612

ABSTRACT

The identification of Mincle as the C-type lectin receptor on innate immune cells responsible for binding TDM and the realization that this receptor could be key to productive vaccines for mycobacterial infection has raised interest in the development of synthetic Mincle ligands as novel adjuvants. We recently reported on the synthesis and evaluation of Brartemicin analog UM-1024 that demonstrated Mincle agonist activity, exhibiting potent Th1/Th17 adjuvant activity that was greater than that of trehalose dibehenate (TDB). Our pursuit to understand Mincle/ligand relationships and improve the pharmacologic properties of the ligands has expanded and continues to reveal new and exciting structure activity relationships. Herein we report the synthesis of novel bi-aryl trehalose derivatives in good to excellent yields. These compounds were evaluated for their ability to engage the human Mincle receptor and tested for the induction of cytokines from human peripheral blood mononuclear cells. A preliminary structure-activity relationship (SAR) of these novel bi-aryl derivatives revealed that bi-aryl trehalose ligand 3D showed relatively high potency in cytokine production in comparison to trehalose glycolipid adjuvant TDB and the natural ligand TDM and induced dose-dependent, Mincle selective stimulation in hMincle HEK reporter cells. Also, through computational studies, we provide an insight into the potential mode of binding of 6,6'-Biaryl trehalose compounds on human Mincle receptor.

4.
ChemMedChem ; 16(8): 1246-1251, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33415819

ABSTRACT

Mincle agonists have been shown to induce inflammatory cytokine production, such as tumor necrosis factor-alpha (TNF) and promote the development of a Th1/Th17 immune response that might be crucial to development of effective vaccination against pathogens such as Mycobacterium tuberculosis. As an expansion of our previous work, a library of 6,6'-amide and sulfonamide α,α-d-trehalose compounds with various substituents on the aromatic ring was synthesized efficiently in good to excellent yields. These compounds were evaluated for their ability to activate the human C-type lectin receptor Mincle by the induction of cytokines from human peripheral blood mononuclear cells. A preliminary structure-activity relationship (SAR) of these novel trehalose diamides and sulfonamides revealed that aryl amide-linked trehalose compounds demonstrated improved activity and relatively high potency cytokine production compared to the Mincle ligand trehalose dibehenate adjuvant (TDB) and the natural ligand trehalose dimycolate (TDM) inducing dose-dependent and human-Mincle-specific stimulation in a HEK reporter cell line.


Subject(s)
Adjuvants, Immunologic/pharmacology , Lectins, C-Type/agonists , Receptors, Immunologic/agonists , Sulfonamides/pharmacology , Trehalose/analogs & derivatives , Trehalose/pharmacology , Adjuvants, Immunologic/chemical synthesis , HEK293 Cells , Humans , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/pharmacology , Sulfonamides/chemical synthesis , Tumor Necrosis Factor-alpha/metabolism
5.
Bioorg Med Chem ; 28(14): 115564, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32616186

ABSTRACT

6,6'-Aryl trehalose derivatives have been synthesized with a view towards identifying novel Th-17-inducing vaccine adjuvants based on the high affinity Mincle ligand Brartemicin. The initial structure-activity relationships of these novel trehalose-based compounds were investigated. All compounds have been evaluated for their ability to engage the Mincle receptor and induce a potential pro-Th17 cytokine profile from human peripheral blood mononuclear cells based on IL-6 production in human peripheral blood mononuclear cells. The preliminary biological characterization of the designed analogs presented in this paper should aid in the future design and testing of more affine ligands that may foster the discovery of novel adjuvants with improved pharmacological properties.


Subject(s)
Lectins, C-Type/metabolism , Receptors, Immunologic/metabolism , Trehalose/pharmacology , Dose-Response Relationship, Drug , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Ligands , Molecular Structure , Structure-Activity Relationship , Trehalose/analogs & derivatives , Trehalose/chemistry
6.
J Med Chem ; 63(1): 309-320, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31809053

ABSTRACT

Mycobacterium tuberculosis (Mtb) continues to be a major health threat worldwide, and the development of Mtb vaccines could play a pivotal role in the prevention and control of this devastating epidemic. Th17-mediated immunity has been implicated in disease protection correlates of immune protection against Mtb. Currently, there are no approved adjuvants capable of driving a Th17 response in a vaccine setting. Recent clinical trial results using trehalose dibehenate have demonstrated a formulation-dependant proof of concept adjuvant system CAF01 capable of inducing long-lived protection. We have discovered a new class of Th17-inducing vaccine adjuvants based on the natural product Brartemicin. We synthesized and evaluated the capacity of a library of aryl trehalose derivatives to drive immunostimulatory reresponses and evaluated the structure-activity relationships in terms of the ability to engage the Mincle receptor and induce production of innate cytokines from human and murine cells. We elaborated on the structure-activity relationship of the new scaffold and demonstrated the ability of the lead entity to induce a pro-Th17 cytokine profile from primary human peripheral blood mononuclear cells and demonstrated efficacy in generating antibodies in combination with tuberculosis antigen M72 in a mouse model.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Mycobacterium tuberculosis/drug effects , Trehalose/analogs & derivatives , Adjuvants, Immunologic/chemical synthesis , Adjuvants, Immunologic/metabolism , Animals , Binding Sites , Cattle , Cell Line , Female , Humans , Lectins, C-Type/agonists , Lectins, C-Type/metabolism , Mice, Inbred BALB C , Molecular Docking Simulation , Molecular Structure , Receptors, Immunologic/agonists , Receptors, Immunologic/metabolism , Structure-Activity Relationship , Trehalose/chemical synthesis , Trehalose/metabolism , Trehalose/therapeutic use , Tuberculosis/therapy , Tuberculosis Vaccines/therapeutic use
7.
Front Immunol ; 10: 338, 2019.
Article in English | MEDLINE | ID: mdl-30873180

ABSTRACT

Despite the ever present need for an effective Mycobacterium tuberculosis (Mtb) vaccine, efforts for development have been largely unsuccessful. Correlates of immune protection against Mtb are not wholly defined, but Th1 and likely Th17 adaptive immune responses have been demonstrated to be necessary for vaccine-mediated protection. Unfortunately, no approved adjuvants are able to drive a Th17 response, though recent clinical trials with CAF01 have demonstrated proof of concept. Herein we present the discovery and characterization of a new class of potential Th17-inducing vaccine adjuvants, alpha-branched trehalose diester molecules (αTDE). Based off the Mtb immunostimulatory component trehalose dimycolate (TDM), we synthesized and evaluated the immunostimulatory capacity of a library of structural derivatives. We evaluated the structure activity relationship of the compounds in relation to chain length and engagement of the Mincle receptor, production of innate cytokines from human and murine cells, and a pro-Th17 cytokine profile from primary human peripheral blood mononuclear cells. Murine cells displayed more structural tolerance, engaging and responding to a wide array of compound chain lengths. Interestingly, human cells displayed a unique specificity for ester chains between 5 and 14 carbons for maximal immune stimulating activity. Evaluation of two distinct αTDEs, B16 and B42, in concert with a recombinant Mtb antigen demonstrated their ability to augment a Th17 immune response against a Mtb antigen in vivo. Collectively this data describes the species-specific structural requirements for maximal human activity of alpha-branched trehalose diester compounds and demonstrates their capacity to serve as potent Th17-inducing adjuvants.


Subject(s)
Cord Factors/chemistry , Cord Factors/immunology , Trehalose/chemistry , Trehalose/immunology , Adjuvants, Immunologic , Animals , Cell Line , Cytokines/metabolism , Humans , Immunity, Cellular , Lectins, C-Type , Mice , Molecular Structure , Mycobacterium tuberculosis/immunology , Structure-Activity Relationship , Th17 Cells/immunology , Th17 Cells/metabolism
8.
Proc Natl Acad Sci U S A ; 99(10): 6901-6, 2002 May 14.
Article in English | MEDLINE | ID: mdl-11983860

ABSTRACT

Phagocytes are a critical component of the innate immune response in humans and eliminate invading microorganisms through a process known as phagocytosis. Two distinct receptor-linked phagocytic pathways, one with Ab receptors (FcRs; FcR, Fc receptor) and the other complement receptors (CRs), mediate binding and ingestion of pathogens by human polymorphonuclear leukocytes (PMNs). Although progress has been made toward defining complex signal transduction processes that underlie phagocytosis in each pathway, very little is known about gene regulation during or after phagocytosis. Therefore, we used human oligonucleotide microarrays to identify changes in expression of 12,561 genes accompanying FcR- and CR-mediated phagocytosis. Eighty-four percent of 279 differentially expressed genes were induced or repressed 90 min after ingestion of Ab- and/or complement-opsonized particles. Unexpectedly, more than 30 of these genes encoded proteins involved in at least three distinct apoptotic pathways. Ninety-four differentially expressed cell fate-related genes were identified between 180 and 360 min after phagocytosis and most were induced or repressed by PMNs activated through both receptors simultaneously. By using flow cytometry, we found that FcR- and CR-mediated phagocytosis each promoted programmed cell death in human PMNs; however, phagocytosis mediated by the combination of FcRs and CRs induced apoptosis earlier than that by either receptor alone. Our results reveal distinct patterns of receptor-mediated gene expression that define complex inducible apoptotic pathways in activated PMNs. Most significantly, we discovered that programmed cell death is regulated at the level of gene expression. Thus, we hypothesize that gene regulation in PMNs facilitates resolution of inflammatory responses.


Subject(s)
Gene Expression , Neutrophils , Phagocytosis , Receptors, Complement/metabolism , Receptors, Fc/metabolism , Apoptosis , Cell Differentiation , Gene Expression Profiling , Humans , Neutrophils/cytology , Neutrophils/metabolism , Phagocytosis/genetics , Phagocytosis/physiology , Polymerase Chain Reaction/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...