Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Eur J Immunol ; 54(5): e2350715, 2024 May.
Article in English | MEDLINE | ID: mdl-38446066

ABSTRACT

Although a role for TLR2 on T cells has been indicated in prior studies, in vivo stimulation of TLR2 on T cells by Mtb and its impact on Mtb infection has not been tested. Furthermore, it is not known if the enhanced susceptibility to Mtb of Tlr2 gene knockout mice is due to its role in macrophages, T cells, or both. To address TLR2 on T cells, we generated Tlr2fl/flxCd4cre/cre mice, which lack expression of TLR2 on both CD4 and CD8 T cells, to study the in vivo role of TLR2 on T cells after aerosol infection with virulent Mtb. Deletion of TLR2 in CD4+ and CD8+ T cells reduces their ability to be co-stimulated by TLR2 ligands for cytokine production. These include both pro- (IFN-γ, TNF-α) and anti-inflammatory cytokines (IL-10). Deletion of TLR2 in T cells affected control of Mtb in the lungs and spleens of infected mice. This suggests that T-cell co-stimulation by mycobacterial TLR2 ligands in vivo contributes to the control of Mtb infection in the lung and spleen.


Subject(s)
CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Mice, Knockout , Mycobacterium tuberculosis , Toll-Like Receptor 2 , Tuberculosis , Animals , Toll-Like Receptor 2/immunology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Mice , CD8-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Tuberculosis/microbiology , Mice, Inbred C57BL , Lung/immunology , Lung/microbiology , Spleen/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Lymphocyte Activation/immunology , Cytokines/metabolism , Cytokines/immunology
2.
Infect Immun ; 89(1)2020 12 15.
Article in English | MEDLINE | ID: mdl-33077627

ABSTRACT

This study investigated responses to Toll-like receptor 2 (TLR2)-driven extracellular signal-related kinase (ERK) signaling in dendritic cells (DCs) versus macrophages. TLR2 signaling was induced with Pam3Cys-Ser-Lys4, and the role of ERK signaling was interrogated pharmacologically with MEK1/2 inhibitor U0126 or genetically with bone marrow-derived macrophages or DCs from Tpl2-/- mice. We assessed cytokine production via enzyme-linked immunosorbent assay (ELISA) or V-Plex, and mRNA levels were assessed via reverse transcriptase quantitative PCR (qRT-PCR). In macrophages, blockade of ERK signaling by pharmacologic or genetic approaches inhibited interleukin 10 (IL-10) expression and increased expression of the p40 subunit shared by IL-12 and IL-23 (IL-12/23p40). In DCs, blockade of ERK signaling similarly inhibited IL-10 expression but decreased IL-12/23p40 expression, which is opposite to the effect of ERK signaling blockade on IL-12/23p40 in macrophages. This difference in IL-12/23p40 regulation correlated with the differential expression of transcription factors cFos and IRF1, which are known to regulate IL-12 family members, including IL-12 and IL-23. Thus, the impact of ERK signaling in response to TLR2 stimulation differs between macrophages and DCs, potentially regulating their distinctive functions in the immune system. ERK-mediated suppression of IL-12/23p40 in macrophages may prevent excessive inflammation and associated tissue damage following TLR2-stimulation, while ERK-mediated induction of IL-12/23p40 in DCs may promote priming of T helper 1 (Th1) responses. A greater understanding of the role that ERK signaling plays in different immune cell types may inform the development of host-directed therapy and optimal adjuvanticity for a number of infectious pathogens.


Subject(s)
Dendritic Cells/metabolism , Interleukin-12/metabolism , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System , Macrophages/metabolism , Proto-Oncogene Proteins/metabolism , Toll-Like Receptor 2/metabolism , Animals , Arginase/genetics , Arginase/metabolism , Dendritic Cells/immunology , Disease Models, Animal , Gene Expression Regulation , Interleukin-10/metabolism , MAP Kinase Kinase Kinases/genetics , Macrophages/immunology , Mice , Mice, Knockout , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Proto-Oncogene Proteins/genetics
3.
J Immunol ; 198(5): 2028-2037, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28122965

ABSTRACT

Mycobacterium tuberculosis utilizes multiple mechanisms to evade host immune responses, and inhibition of effector CD4+ T cell responses by M. tuberculosis may contribute to immune evasion. TCR signaling is inhibited by M. tuberculosis cell envelope lipoglycans, such as lipoarabinomannan and lipomannan, but a mechanism for lipoglycans to traffic from M. tuberculosis within infected macrophages to reach T cells is unknown. In these studies, we found that membrane vesicles produced by M. tuberculosis and released from infected macrophages inhibited the activation of CD4+ T cells, as indicated by reduced production of IL-2 and reduced T cell proliferation. Flow cytometry and Western blot demonstrated that lipoglycans from M. tuberculosis-derived bacterial vesicles (BVs) are transferred to T cells, where they inhibit T cell responses. Stimulation of CD4+ T cells in the presence of BVs induced expression of GRAIL, a marker of T cell anergy; upon restimulation, these T cells showed reduced ability to proliferate, confirming a state of T cell anergy. Furthermore, lipoarabinomannan was associated with T cells after their incubation with infected macrophages in vitro and when T cells were isolated from lungs of M. tuberculosis-infected mice, confirming the occurrence of lipoarabinomannan trafficking to T cells in vivo. These studies demonstrate a novel mechanism for the direct regulation of CD4+ T cells by M. tuberculosis lipoglycans conveyed by BVs that are produced by M. tuberculosis and released from infected macrophages. These lipoglycans are transferred to T cells to inhibit T cell responses, providing a mechanism that may promote immune evasion.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immune Evasion , Lung/immunology , Macrophages/immunology , Mycobacterium tuberculosis/immunology , Secretory Vesicles/microbiology , Tuberculosis/immunology , Animals , Cell Proliferation , Cells, Cultured , Clonal Anergy , Female , Humans , Lipopolysaccharides/immunology , Lung/microbiology , Lymphocyte Activation , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Secretory Vesicles/immunology
4.
PLoS One ; 10(10): e0140064, 2015.
Article in English | MEDLINE | ID: mdl-26445168

ABSTRACT

Macrophages depend on colony stimulating factor 1 (also known as M-CSF) for their growth and differentiation, but the requirements for intracellular signals that lead to macrophage differentiation and function remain unclear. M-CSF is known to activate ERK1 and ERK2, but the importance of this signaling pathway in macrophage development is unknown. In these studies, we characterized a novel model of Erk1(-/-) Erk2(flox/flox) Lyz2(Cre/Cre) mice in which the ERK2 isoform is deleted from macrophages in the background of global ERK1 deficiency. Cultures of M-CSF-stimulated bone marrow precursors from these mice yielded reduced numbers of macrophages. Whereas macrophages developing from M-CSF-stimulated bone marrow of Erk2(flox/flox) Lyz2(Cre/Cre) mice showed essentially complete loss of ERK2 expression, the reduced number of macrophages that develop from Erk1(-/-) Erk2(flox/flox) Lyz2(Cre/Cre) bone marrow show retention of ERK2 expression, indicating selective outgrowth of a small proportion of precursors in which Cre-mediated deletion failed to occur. The bone marrow of Erk1(-/-) Erk2(flox/flox) Lyz2(Cre/Cre) mice was enriched for CD11b+ myeloid cells, CD11b(hi) Gr-1(hi) neutrophils, Lin- c-Kit+ Sca-1+ hematopoietic stem cells, and Lin- c-Kit+ CD34+ CD16/32+ granulocyte-macrophage progenitors. Culture of bone marrow Lin- cells under myeloid-stimulating conditions yielded reduced numbers of monocytes. Collectively, these data indicate that the defect in production of macrophages is not due to a reduced number of progenitors, but rather due to reduced ability of progenitors to proliferate and produce macrophages in response to M-CSF-triggered ERK signaling. Macrophages from Erk1(-/-) Erk2(flox/flox) Lyz2(Cre/Cre) bone marrow showed reduced induction of M-CSF-regulated genes that depend on the ERK pathway for their expression. These data demonstrate that ERK1/ERK2 play a critical role in driving M-CSF-dependent proliferation of bone marrow progenitors for production of macrophages.


Subject(s)
Bone Marrow Cells/cytology , MAP Kinase Signaling System , Macrophages/cytology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Animals , Bone Marrow Cells/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Gene Deletion , Gene Expression Regulation , Granulocytes/cytology , Granulocytes/metabolism , Hematopoiesis , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Monocytes/metabolism
5.
Antimicrob Agents Chemother ; 59(3): 1512-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25534736

ABSTRACT

The dwindling repertoire of antibiotics to treat methicillin-resistant Staphylococcus aureus (MRSA) calls for novel treatment options. Quorum-quenching agents offer an alternative or an adjuvant to antibiotic therapy. Three biaryl hydroxyketone compounds discovered previously (F1, F12, and F19; G. Yu, D. Kuo, M. Shoham, and R. Viswanathan, ACS Comb Sci 16:85-91, 2014) were tested for efficacy in MRSA-infected animal models. Topical therapy of compounds F1 and F12 in a MRSA murine wound infection model promotes wound healing compared to the untreated control. Compounds F1, F12, and F19 afford significant survival benefits in a MRSA insect larva model. Combination therapy of these quorum-quenching agents with cephalothin or nafcillin, antibiotics to which MRSA is resistant in monotherapy, revealed additional survival benefits. The quorum-quenching agents sensitize MRSA to the antibiotic by a synergistic mode of action that also is observed in vitro. An adjuvant of 1 µg/ml F1, F12, or F19 reduces the MIC of nafcillin and cephalothin about 50-fold to values comparable to those for vancomycin, the antibiotic often prescribed for MRSA infections. These findings suggest that it is possible to resurrect obsolete antibiotic therapies in combination with these novel quorum-quenching agents.


Subject(s)
Anti-Bacterial Agents/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Quorum Sensing/drug effects , Wound Healing/drug effects , beta-Lactams/pharmacology , Animals , Cell Line , Cephalothin/pharmacology , Macrophages/immunology , Mice , Microbial Sensitivity Tests , Nafcillin/pharmacology
6.
Eur J Immunol ; 44(5): 1410-21, 2014 May.
Article in English | MEDLINE | ID: mdl-24497180

ABSTRACT

We have previously demonstrated that mycobacterial lipoproteins engage TLR2 on human CD4(+) T cells and upregulate TCR-triggered IFN-γ secretion and cell proliferation in vitro. Here we examined the role of CD4(+) T-cell-expressed TLR2 in Mycobacterium tuberculosis (MTB) Ag-specific T-cell priming and in protection against MTB infection in vivo. Like their human counterparts, mouse CD4(+) T cells express TLR2 and respond to TLR2 costimulation in vitro. This Th1-like response was observed in the context of both polyclonal and Ag-specific TCR stimulation. To evaluate the role of T-cell TLR2 in priming of CD4(+) T cells in vivo, naive MTB Ag85B-specific TCR transgenic CD4(+) T cells (P25 TCR-Tg) were adoptively transferred into Tlr2(-/-) recipient C57BL/6 mice that were then immunized with Ag85B and with or without TLR2 ligand Pam3 Cys-SKKKK. TLR2 engagement during priming resulted in increased numbers of IFN-γ-secreting P25 TCR-Tg T cells 1 week after immunization. P25 TCR-Tg T cells stimulated in vitro via TCR and TLR2 conferred more protection than T cells stimulated via TCR alone when adoptively transferred before MTB infection. Our findings indicate that TLR2 engagement on CD4(+) T cells increases MTB Ag-specific responses and may contribute to protection against MTB infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Mycobacterium tuberculosis/immunology , Toll-Like Receptor 2/immunology , Tuberculosis/immunology , Acyltransferases/biosynthesis , Acyltransferases/genetics , Acyltransferases/immunology , Acyltransferases/pharmacology , Animals , Antigens, Bacterial/biosynthesis , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Antigens, Bacterial/pharmacology , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Proteins/pharmacology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Chromobox Protein Homolog 5 , Humans , Immunization , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interferon-gamma/immunology , Mice , Mice, Knockout , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Toll-Like Receptor 2/biosynthesis , Toll-Like Receptor 2/genetics , Tuberculosis/genetics , Tuberculosis/metabolism , Tuberculosis/pathology , Tuberculosis/prevention & control
7.
Cell Immunol ; 254(2): 94-104, 2009.
Article in English | MEDLINE | ID: mdl-18762288

ABSTRACT

Mycobacterium tuberculosis and M. bovis BCG infect APCs. In vitro, mycobacteria inhibit IFN-gamma-induced MHC-II expression by macrophages, but the effects of mycobacteria on lung APCs in vivo remain unclear. To assess MHC-II expression on APCs infected in vivo, mice were aerosol-infected with GFP-expressing BCG. At 28 d, approximately 1% of lung APCs were GFP+ by flow cytometry and CFU data. Most GFP+ cells were CD11b(high)/CD11c(neg-mid) lung macrophages (58-68%) or CD11b(high)/CD11c(high) DCs (28-31%). Lung APC MHC-II expression was higher in infected mice than naïve mice. Within infected lungs, however, MHC-II expression was lower in GFP+ cells than GFP- cells for both macrophages and DCs. MHC-II expression was also inhibited on purified lung macrophages and DCs that were infected with BCG in vitro. Thus, lung APCs that harbor mycobacteria in vivo have decreased MHC-II expression relative to uninfected APCs from the same lung, possibly contributing to evasion of T cell responses.


Subject(s)
Dendritic Cells/immunology , Histocompatibility Antigens Class II/immunology , Macrophages, Alveolar/immunology , Mycobacterium bovis/immunology , Tuberculosis/immunology , Animals , Antigen-Presenting Cells/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , CD11 Antigens/immunology , Cell Differentiation/immunology , Female , Macrophages, Alveolar/cytology , Mice , Mice, Inbred C57BL
8.
Biochemistry ; 45(11): 3733-9, 2006 Mar 21.
Article in English | MEDLINE | ID: mdl-16533056

ABSTRACT

Abnormal carbohydrates expressed on tumor cells, which are termed tumor-associated carbohydrate antigens (TACAs), are potential targets for the development of cancer vaccines. However, immune tolerance to TACAs has severely hindered progress in this area. To overcome this problem, we have developed a novel immunotherapeutic strategy based on synthetic cancer vaccines and metabolic engineering of TACAs on tumor cells. One critical step of this new strategy is metabolic engineering of cancer, namely, to induce expression of an artificial form of a TACA by supplying tumors with an artificial monosaccharide precursor. To identify the proper precursor for this application, N-propionyl, N-butanoyl, N-isobutanoyl, and N-phenylacetyl derivatives of d-mannosamine were synthesized, and their efficiency as biosynthetic precursors in modifying sialic acid and inducing expression of modified forms of GM3 antigen on tumor cells was investigated. For this purpose, tumor cells were incubated with different N-acyl-d-mannosamines, and modified forms of GM3 expressed on tumor cells were analyzed by flow cytometry using antigen-specific antisera. N-Phenylacetyl-d-mannosamine was efficiently incorporated in a time- and dose-dependent manner to bioengineer GM3 expression by several tumor cell lines, including K562, SKMEL-28, and B16-F0. Moreover, these tumor cell lines also exhibited ManPAc-dependent sensitivity to cytotoxicity mediated by anti-PAcGM3 immune serum and complement. These results provide an important validation for this novel therapeutic strategy. Because N-phenylacetyl GM3-protein conjugates are particularly immunogenic, the combination of an N-phenylacetyl GM3 conjugate vaccine with systemic N-phenylacetyl-d-mannosamine treatment is a promising immunotherapy for future development and application to melanoma and other GM3-bearing tumors.


Subject(s)
Antigens, Tumor-Associated, Carbohydrate/metabolism , G(M3) Ganglioside/chemistry , Hexosamines/chemical synthesis , Hexosamines/pharmacology , Animals , Antigens, Tumor-Associated, Carbohydrate/pharmacology , Biochemistry/methods , Cancer Vaccines/biosynthesis , Carbohydrate Sequence , Dose-Response Relationship, Drug , Flow Cytometry , G(M3) Ganglioside/immunology , Glycoconjugates/metabolism , Glycoconjugates/pharmacology , Hexosamines/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , Molecular Sequence Data , N-Acetylneuraminic Acid/metabolism , Time Factors , Tumor Cells, Cultured
9.
J Med Chem ; 48(3): 875-83, 2005 Feb 10.
Article in English | MEDLINE | ID: mdl-15689172

ABSTRACT

The problem of immunotolerance to GM3, an important tumor-associated trisaccharide antigen, seriously hinders its usage in cancer vaccine development. To solve this problem, the keyhole limpet hemocyanin (KLH) conjugates of a series of GM3 derivatives were synthesized and screened as therapeutic cancer vaccines. First, the beta-linked anomeric azides of differently N-acylated GM3 analogues were prepared by a highly convergent procedure. Next, a pentenoyl group was linked to the reducing end of the carbohydrate antigens following selective reduction of the azido group. The linker was thereafter ozonolyzed to give an aldehyde functionality permitting the conjugation of the antigens to KLH via reductive amination. Finally, the immunological properties of the resultant glycoconjugates were studied in C57BL/6 mice by assessing the titers of specific antibodies induced by the GM3 analogues. While KLH-GM3 elicited low levels of immune response, the KLH conjugates of N-propionyl, N-butanoyl, N-iso-butanoyl, and N-phenylacetyl GM3s induced robust immune reactions with antibodies of multiple isotypes, indicating significantly improved and T-cell dependent immune responses that lead to isotype switching, affinity maturation, and the induction of immunological "memory". It was suggested that GM3PhAc-KLH is a promising vaccine candidate for glycoengineered immunotherapy of cancer with GM3 as the primary target.


Subject(s)
Cancer Vaccines/chemical synthesis , Glycoconjugates/chemical synthesis , Hemocyanins/chemistry , Immunoglobulin Gm Allotypes/chemistry , Animals , Cancer Vaccines/immunology , Carbohydrate Sequence , Enzyme-Linked Immunosorbent Assay , Female , Glycoconjugates/immunology , Immune Tolerance , Immunoglobulin G/blood , Immunoglobulin Gm Allotypes/immunology , Immunoglobulin M/blood , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Structure-Activity Relationship , Vaccines, Conjugate/immunology
10.
Glycoconj J ; 20(6): 407-14, 2004.
Article in English | MEDLINE | ID: mdl-15238705

ABSTRACT

The overexpression of N -acetylneuraminic acid (Neu5Ac) is closely correlated with malignant transformations. Thus, Neu5Ac is an important target in the design of cancer vaccines. To study the influence of chemical modifications of Neu5Ac on its immunological properties, the alpha-allyl glycosides of five differently N -acylated neuraminic acid derivatives were prepared. Following selective ozonolysis of their allyl group to form an aldehyde functionality, they were coupled to keyhole limpet hemocyanin (KLH) via reductive amination. Resultant glycoconjugates were studied in C57BL/6 mice. The N -propionyl, N - iso- butanoyl and N -phenylacetyl derivatives of neuraminic acid provoked robust immune responses of various antibody isotypes, including IgM, IgG1, IgG2a and IgG3, whereas N -trifluoropropionylneuraminic acid and natural Neu5Ac were essentially nonimmunogenic. Moreover, the N -phenylacetyl and N - iso- butanoyl derivatives mainly induced IgG responses that are desirable for antitumor applications. These results raise the promise of formulating effective glycoconjugate cancer vaccines via derivatizing sialic acid residues of sialooligosaccharides.


Subject(s)
Neuraminic Acids/chemistry , Neuraminic Acids/chemical synthesis , Animals , Cancer Vaccines/chemistry , Carbohydrate Sequence , Cell Transformation, Neoplastic , Enzyme-Linked Immunosorbent Assay , Female , Glycoconjugates/chemistry , Hemocyanins/chemistry , Immunoglobulin G/chemistry , Immunoglobulin M/chemistry , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred C57BL , Models, Chemical , Neuraminic Acids/immunology , Oligosaccharides/chemistry , Ozone , Sialic Acids/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...