Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Pharmaceutics ; 15(12)2023 Nov 21.
Article in English | MEDLINE | ID: mdl-38139993

ABSTRACT

Pancreatic cancer remains a formidable challenge due to limited treatment options and its aggressive nature. In recent years, the naturally occurring anticancer compound juglone has emerged as a potential therapeutic candidate, showing promising results in inhibiting tumor growth and inducing cancer cell apoptosis. However, concerns over its toxicity have hampered juglone's clinical application. To address this issue, we have explored the use of polymeric micelles as a delivery system for juglone in pancreatic cancer treatment. These micelles, formulated using Poloxamer 407 and D-α-Tocopherol polyethylene glycol 1000 succinate, offer an innovative solution to enhance juglone's therapeutic potential while minimizing toxicity. In-vitro studies have demonstrated that micelle-formulated juglone (JM) effectively decreases proliferation and migration and increases apoptosis in pancreatic cancer cell lines. Importantly, in-vivo, JM exhibited no toxicity, allowing for increased dosing frequency compared to free drug administration. In mice, JM significantly reduced tumor growth in subcutaneous xenograft and orthotopic pancreatic cancer models. Beyond its direct antitumor effects, JM treatment also influenced the tumor microenvironment. In immunocompetent mice, JM increased immune cell infiltration and decreased stromal deposition and activation markers, suggesting an immunomodulatory role. To understand JM's mechanism of action, we conducted RNA sequencing and subsequent differential expression analysis on tumors that were treated with JM. The administration of JM treatment reduced the expression levels of the oncogenic protein MYC, thereby emphasizing its potential as a focused, therapeutic intervention. In conclusion, the polymeric micelles-mediated delivery of juglone holds excellent promise in pancreatic cancer therapy. This approach offers improved drug delivery, reduced toxicity, and enhanced therapeutic efficacy.

2.
Nat Commun ; 14(1): 5665, 2023 09 13.
Article in English | MEDLINE | ID: mdl-37704631

ABSTRACT

Triple-negative breast cancer (TNBC) patients have a poor prognosis and few treatment options. Mouse models of TNBC are important for development of new therapies, however, few mouse models represent the complexity of TNBC. Here, we develop a female TNBC murine model by mimicking two common TNBC mutations with high co-occurrence: amplification of the oncogene MYC and deletion of the tumor suppressor PTEN. This Myc;Ptenfl model develops heterogeneous triple-negative mammary tumors that display histological and molecular features commonly found in human TNBC. Our research involves deep molecular and spatial analyses on Myc;Ptenfl tumors including bulk and single-cell RNA-sequencing, and multiplex tissue-imaging. Through comparison with human TNBC, we demonstrate that this genetic mouse model develops mammary tumors with differential survival and therapeutic responses that closely resemble the inter- and intra-tumoral and microenvironmental heterogeneity of human TNBC, providing a pre-clinical tool for assessing the spectrum of patient TNBC biology and drug response.


Subject(s)
Mammary Neoplasms, Animal , Triple Negative Breast Neoplasms , Animals , Female , Humans , Mice , Aggression , Disease Models, Animal , Mutation , PTEN Phosphohydrolase/genetics , Triple Negative Breast Neoplasms/genetics , Proto-Oncogene Proteins c-myc/metabolism
3.
PLoS Pathog ; 19(1): e1010351, 2023 01.
Article in English | MEDLINE | ID: mdl-36689486

ABSTRACT

CMV, a ubiquitous herpesvirus, elicits an extraordinarily large T cell response that is sustained or increases over time, a phenomenon termed 'memory inflation.' Remarkably, even latent, non-productive infection can drive memory inflation. Despite intense research on this phenomenon, the infected cell type(s) involved are unknown. To identify the responsible cell type(s), we designed a Cre-lox murine CMV (MCMV) system, where a spread-deficient (ΔgL) virus expresses recombinant SIINFEKL only in Cre+ host cells. We found that latent infection of endothelial cells (ECs), but not dendritic cells (DCs) or hepatocytes, was sufficient to drive CD8 T cell memory inflation. Infection of Lyve-1-Cre and Prox1-CreERT2 mice revealed that amongst EC subsets, infection of lymphatic ECs was sufficient. Genetic ablation of ß2m on lymphatic ECs did not prevent inflation, suggesting another unidentified cell type can also present antigen to CD8 T cells during latency. This novel system definitively shows that antigen presentation by lymphatic ECs drives robust CD8 T cell memory inflation.


Subject(s)
Cytomegalovirus Infections , Latent Infection , Muromegalovirus , Animals , Mice , Endothelial Cells , CD8-Positive T-Lymphocytes , Antigens , Immunologic Memory
4.
Mol Cancer Res ; 20(7): 1151-1165, 2022 07 06.
Article in English | MEDLINE | ID: mdl-35380701

ABSTRACT

As a transcription factor that promotes cell growth, proliferation, and apoptosis, c-MYC (MYC) expression in the cell is tightly controlled. Disruption of oncogenic signaling pathways in human cancers can increase MYC protein stability, due to altered phosphorylation ratios at two highly conserved sites, Threonine 58 (T58) and Serine 62 (S62). The T58 to Alanine mutant (T58A) of MYC mimics the stabilized, S62 phosphorylated, and highly oncogenic form of MYC. The S62A mutant is also stabilized, lacks phosphorylation at both Serine 62 and Threonine 58, and has been shown to be nontransforming in vitro. However, several regulatory proteins are reported to associate with MYC lacking phosphorylation at S62 and T58, and the role this form of MYC plays in MYC transcriptional output and in vivo oncogenic function is understudied. We generated conditional c-Myc knock-in mice in which the expression of wild-type MYC (MYCWT), the T58A mutant (MYCT58A), or the S62A mutant (MYCS62A) with or without expression of endogenous Myc is controlled by the T-cell-specific Lck-Cre recombinase. MYCT58A expressing mice developed clonal T-cell lymphomas with 100% penetrance and conditional knock-out of endogenous Myc accelerated this lymphomagenesis. In contrast, MYCS62A mice developed clonal T-cell lymphomas at a much lower penetrance, and the loss of endogenous MYC reduced the penetrance while increasing the appearance of a non-transgene driven B-cell lymphoma with splenomegaly. Together, our study highlights the importance of regulated phosphorylation of MYC at T58 and S62 for T-cell transformation. IMPLICATIONS: Dysregulation of phosphorylation at conserved T58 and S62 residues of MYC differentially affects T-cell development and lymphomagenesis.


Subject(s)
Lymphoma, T-Cell , Proto-Oncogene Proteins c-myc , Threonine , Animals , Carcinogenesis , Mice , Phosphorylation , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Serine/metabolism , T-Lymphocytes/metabolism , Threonine/genetics , Transcription Factors/metabolism
5.
Eur J Immunol ; 51(6): 1473-1481, 2021 06.
Article in English | MEDLINE | ID: mdl-33684223

ABSTRACT

Therapeutic antibodies blocking PD-1-/PD-L1 interaction have achieved remarkable clinical success in cancer. In addition to blocking a target molecule, some isotypes of antibodies can activate complement, NK cells or phagocytes, resulting in death of the cell expressing the antibody's target. Human anti-PD-1 therapeutics use antibody isotypes designed to minimize such antibody-dependent lysis. In contrast, anti-PD-1 reagents used in mice are derived from multiple species, with different isotypes, and are not engineered to reduce target cell death: few studies analyze or discuss how antibody species and isotype may impact data interpretation. We demonstrate here that anti-PD-1 therapy to promote activation and proliferation of murine PD-1-expressing CD8 T cells sometimes led instead to a loss of antigen specific cells. This phenomenon was seen in two tumor models and a model of virus infection, and varied with the clone of anti-PD-1 antibody. Additionally, we compared competition among anti-PD-1 clones to find a combination that allows detection of PD-1-expressing cells despite the presence of blocking anti-PD1 antibodies in vivo. These data bring attention to the possibility of unintended target cell depletion with some commonly used anti-mouse PD-1 clones, and should provide a valuable resource for the design and interpretation of anti-PD-1 studies in mice.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Herpesviridae Infections/immunology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Muromegalovirus/physiology , Sarcoma/immunology , Skin Neoplasms/immunology , Animals , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/transplantation , Cell Death , Cell Line, Tumor , Cricetinae , Disease Models, Animal , Drug Evaluation, Preclinical , Herpesviridae Infections/therapy , Humans , Immunoglobulin G/metabolism , Immunoglobulin Isotypes/metabolism , Methylcholanthrene , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Rats , Sarcoma/therapy , Skin Neoplasms/therapy
6.
MAbs ; 12(1): 1834818, 2020.
Article in English | MEDLINE | ID: mdl-33151105

ABSTRACT

Simulating a viral infection in tumor cells is an attractive concept to eliminate tumor cells. We previously reported the molecular design and the in vitro potency of recombinant monoclonal antibodies fused to a virus-derived peptide MHC class I complex that bypass the peptide processing and MHC loading pathway and directly displays a viral peptide in an MHC class I complex on the tumor cell surface. Here, we show that a vaccination-induced single peptide-specific CD8 T cell response was sufficient to eliminate B16 melanoma tumor cells in vivo in a fully immunocompetent, syngeneic mouse tumor model when mice were treated with mouse pMHCI-IgGs fusion proteins targeting the mouse fibroblast activation protein. Tumor growth of small, established B16 lung metastases could be controlled. The pMHCI-IgG had similar potency as an analogous pan-CD3 T-cell bispecific antibody. In contrast to growth control of small tumors, none of the compounds controlled larger solid tumors of MC38 cancer cells, despite penetration of pMHCI-IgGs into the tumor tissue and clear attraction and activation of antigen-specific CD8 T cells inside the tumor. pMHCI-IgG can have a similar potency as classical pan-T-cell recruiting molecules. The results also highlight the need to better understand immune suppression in advanced solid tumors.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Histocompatibility Antigens Class I/immunology , Immunoglobulin G/immunology , Melanoma, Experimental/immunology , Animals , Mice , Mice, Inbred C57BL , Recombinant Fusion Proteins/immunology
7.
Sci Immunol ; 2(16)2017 10 13.
Article in English | MEDLINE | ID: mdl-29030501

ABSTRACT

Trafficking of memory CD8+ T cells out of the circulation is essential to provide protective immunity against intracellular pathogens in nonlymphoid tissues. However, the molecular mechanisms that dictate the trafficking potential of diverse memory CD8+ T cell populations are not completely defined. We show that after infection or inflammatory challenge, central memory (TCM) CD8+ T cells rapidly traffic into nonlymphoid tissues, whereas most effector memory cells remain in the circulation. Furthermore, we demonstrate that cellular migration of memory CD8+ T cells into nonlymphoid tissues is driven by interleukin-15 (IL-15)-stimulated enzymatic synthesis of core 2 O-glycans, which generates functional ligands for E- and P-selectins. Given that IL-15-stimulated expression of glycosyltransferase enzymes is largely a feature of TCM CD8+ T cells, this allows TCM to selectively migrate out of the circulation and into nonlymphoid tissues. Collectively, our data indicate that entry of memory CD8+ T cells into inflamed, nonlymphoid tissues is primarily restricted to TCM cells that have the capacity to synthesize core 2 O-glycans.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Polysaccharides/immunology , Animals , CD8-Positive T-Lymphocytes/enzymology , Cell Movement , Cytoplasm/immunology , Cytoplasm/virology , Inflammation , Interleukin-15/genetics , Interleukin-15/immunology , Lymphocytic choriomeningitis virus/immunology , Mice , Polysaccharides/biosynthesis
8.
Vet Immunol Immunopathol ; 188: 65-70, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28615129

ABSTRACT

Herpesviruses establish lifelong infections, normally characterized by prolonged periods of latency with intermittent episodes of viral reactivation. Feline herpesvirus-1 (FHV-1) infects domestic cats, and epidemiological studies indicate that many or most domestic cats are exposed to FHV-1, but the strength and longevity of the antibody response to FHV-1 is not fully characterized. Here we describe development of an ELISA, using lysates of cat cells infected with FHV-1, that measure feline antibodies against FHV-1. The assay is sensitive, quantitative and has a large dynamic range. We found that serum anti-FHV-1 antibodies primarily recognize FHV-1 proteins of the Late (L) class and are primarily of the IgG isotype. We then analyzed serum from a cross-sectional cohort of 100 client-owned cats that differed in age, sex and vaccination history. While there was no difference in FHV-1 antibody responses between females and males, antibody levels were significantly increased in older cats in comparison with younger animals (p=0.01). Surprisingly, as the length of time since the most recent vaccination increased, there was no corresponding drop in serum anti-FHV-1 antibody. These data suggest that FHV-1 immunity is very long-lived and support the current recommendation that many cats do not require revaccination against FHV-1 annually.


Subject(s)
Cat Diseases/virology , Herpesviridae Infections/veterinary , Herpesviridae/immunology , Age Factors , Animals , Antibodies, Viral/immunology , Cat Diseases/immunology , Cats/immunology , Cats/virology , Enzyme-Linked Immunosorbent Assay/veterinary , Female , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Immunity, Humoral/immunology , Immunity, Humoral/physiology , Male
9.
J Exp Med ; 214(7): 1889-1899, 2017 Jul 03.
Article in English | MEDLINE | ID: mdl-28566275

ABSTRACT

Cytomegalovirus (CMV)-based vaccines have shown remarkable efficacy in the rhesus macaque model of acquired immune deficiency syndrome, enabling 50% of vaccinated monkeys to clear a subsequent virulent simian immunodeficiency virus challenge. The protective vaccine elicited unconventional CD8 T cell responses that were entirely restricted by MHC II or the nonclassical MHC I molecule, MHC-E. These unconventional responses were only elicited by a fibroblast-adapted rhesus CMV vector with limited tissue tropism; a repaired vector with normal tropism elicited conventional responses. Testing whether these unusual protective CD8 T responses could be elicited in humans requires vaccinating human subjects with a fibroblast-adapted mutant of human CMV (HCMV). In this study, we describe the CD8 T cell responses of human subjects vaccinated with two fibroblast-adapted HCMV vaccines. Most responses were identified as conventional classically MHC I restricted, and we found no evidence for MHC II or HLA-E restriction. These results indicate that fibroblast adaptation alone is unlikely to explain the unconventional responses observed in macaques.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus Vaccines/immunology , Cytomegalovirus/immunology , Fibroblasts/immunology , Amino Acid Sequence , Cell Line , Cell Line, Tumor , Cells, Cultured , Cytomegalovirus/physiology , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Infections/virology , Cytomegalovirus Vaccines/administration & dosage , Cytomegalovirus Vaccines/genetics , Epitopes/immunology , Fibroblasts/virology , Flow Cytometry , Histocompatibility Antigens Class I/immunology , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Humans , K562 Cells , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Male , Microscopy, Fluorescence , Mutation , Vaccination
10.
Vaccine ; 32(30): 3765-71, 2014 Jun 24.
Article in English | MEDLINE | ID: mdl-24856785

ABSTRACT

Preventing losses in vaccine potency due to accidental freezing has recently become a topic of interest for improving vaccines. All vaccines with aluminum-containing adjuvants are susceptible to such potency losses. Recent studies have described excipients that protect the antigen from freeze-induced inactivation, prevent adjuvant agglomeration and retain potency. Although these strategies have demonstrated success, they do not provide a mechanistic understanding of freeze-thaw (FT) induced potency losses. In the current study, we investigated how adjuvant frozen in the absence of antigen affects vaccine immunogenicity and whether preventing damage to the freeze-sensitive recombinant hepatitis B surface antigen (rHBsAg) was sufficient for maintaining vaccine potency. The final vaccine formulation or Alhydrogel(®) alone was subjected to three FT-cycles. The vaccines were characterized for antigen adsorption, rHBsAg tertiary structure, particle size and charge, adjuvant elemental content and in-vivo potency. Particle agglomeration of either vaccine particles or adjuvant was observed following FT-stress. In vivo studies demonstrated no statistical differences in IgG responses between vaccines with FT-stressed adjuvant and no adjuvant. Adsorption of rHBsAg was achieved; regardless of adjuvant treatment, suggesting that the similar responses were not due to soluble antigen in the frozen adjuvant-containing formulations. All vaccines with adjuvant, including the non-frozen controls, yielded similar, blue-shifted fluorescence emission spectra. Immune response differences could not be traced to differences in the tertiary structure of the antigen in the formulations. Zeta potential measurements and elemental content analyses suggest that FT-stress resulted in a significant chemical alteration of the adjuvant surface. This data provides evidence that protecting a freeze-labile antigen from subzero exposure is insufficient to maintain vaccine potency. Future studies should focus on adjuvant protection. To our knowledge, this is the first study to systematically investigate how FT-stress to adjuvant alone affects immunogenicity. It provides definitive evidence that this damage is sufficient to reduce vaccine potency.


Subject(s)
Adjuvants, Immunologic/chemistry , Aluminum Hydroxide/chemistry , Freezing , Hepatitis B Vaccines/immunology , Vaccine Potency , Animals , Antibodies, Viral/blood , Antibody Formation , Female , Hepatitis B Surface Antigens/chemistry , Hepatitis B Surface Antigens/immunology , Immunoglobulin G/blood , Mice, Inbred BALB C , Particle Size , Protein Structure, Tertiary
11.
Proc Natl Acad Sci U S A ; 110(12): E1122-31, 2013 Mar 19.
Article in English | MEDLINE | ID: mdl-23447566

ABSTRACT

Many vaccines include aluminum salts (alum) as adjuvants despite little knowledge of alum's functions. Host DNA rapidly coats injected alum. Here, we further investigated the mechanism of alum and DNA's adjuvant function. Our data show that DNase coinjection reduces CD4 T-cell priming by i.m. injected antigen + alum. This effect is partially replicated in mice lacking stimulator of IFN genes, a mediator of cellular responses to cytoplasmic DNA. Others have shown that DNase treatment impairs dendritic cell (DC) migration from the peritoneal cavity to the draining lymph node in mice immunized i.p. with alum. However, our data show that DNase does not affect accumulation of, or expression of costimulatory proteins on, antigen-loaded DCs in lymph nodes draining injected muscles, the site by which most human vaccines are administered. DNase does inhibit prolonged T-cell-DC conjugate formation and antigen presentation between antigen-positive DCs and antigen-specific CD4 T cells following i.m. injection. Thus, from the muscle, an immunization site that does not require host DNA to promote migration of inflammatory DCs, alum acts as an adjuvant by introducing host DNA into the cytoplasm of antigen-bearing DCs, where it engages receptors that promote MHC class II presentation and better DC-T-cell interactions.


Subject(s)
Adjuvants, Immunologic/pharmacology , Alum Compounds/pharmacology , Antigen Presentation/drug effects , CD4-Positive T-Lymphocytes/immunology , DNA/immunology , Dendritic Cells/immunology , Histocompatibility Antigens Class II/immunology , Animals , Antigens/immunology , Cell Communication/drug effects , Cell Movement/drug effects , Humans , Mice , Mice, Knockout , Mice, Nude
12.
J Immunol ; 187(3): 1385-92, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21697462

ABSTRACT

NK cells recognize virus-infected cells with germline-encoded activating and inhibitory receptors that do not undergo genetic recombination or mutation. Accordingly, NK cells are often considered part of the innate immune response. The innate response comprises rapid early defenders that do not form immune memory. However, there is increasing evidence that experienced NK cells provide increased protection to secondary infection, a hallmark of the adaptive response. In this study, we compare the dynamics of the innate and adaptive immune responses by examining the kinetic profiles of the NK and T cell response to murine CMV infection. We find that, unexpectedly, the kinetics of NK cell proliferation is neither earlier nor faster than the CD4 or CD8 T cell response. Furthermore, early NK cell contraction after the peak of the response is slower than that of T cells. Finally, unlike T cells, experienced NK cells do not experience biphasic decay after the response peak, a trait associated with memory formation. Rather, NK cell contraction is continuous, constant, and returns to below endogenous preinfection levels. This indicates that the reason why Ag-experienced NK cells remain detectable for a prolonged period after adoptive transfer and infection is in part due to the high precursor frequency, slow decay rate, and low background levels of Ly49H(+) NK cells in recipient DAP12-deficient mice. Thus, the quantitative contribution of Ag-experienced NK cells in an endogenous secondary response, with higher background levels of Ly49H(+) NK cells, may be not be as robust as the secondary response observed in T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Herpesviridae Infections/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Muromegalovirus/immunology , Animals , CD4-CD8 Ratio , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/virology , Herpesviridae Infections/metabolism , Herpesviridae Infections/pathology , Immunologic Memory , Killer Cells, Natural/virology , Liver/immunology , Liver/pathology , Liver/virology , Lung/immunology , Lung/pathology , Lung/virology , Lymphocyte Count , Mice , Mice, Inbred C57BL , Models, Immunological , Spleen/immunology , Spleen/pathology , Spleen/virology , Stem Cells/immunology , Stem Cells/pathology , Stem Cells/virology
13.
Blood ; 116(24): 5191-9, 2010 Dec 09.
Article in English | MEDLINE | ID: mdl-20876456

ABSTRACT

It has been recognized for nearly 80 years that insoluble aluminum salts are good immunologic adjuvants and that they form long-lived nodules in vivo. Nodule formation has long been presumed to be central for adjuvant activity by providing an antigen depot, but the composition and function of these nodules is poorly understood. We show here that aluminum salt nodules formed within hours of injection and contained the clotting protein fibrinogen. Fibrinogen was critical for nodule formation and required processing to insoluble fibrin by thrombin. DNase treatment partially disrupted the nodules, and the nodules contained histone H3 and citrullinated H3, features consistent with extracellular traps. Although neutrophils were not essential for nodule formation, CD11b(+) cells were implicated. Vaccination of fibrinogen-deficient mice resulted in normal CD4 T-cell and antibody responses and enhanced CD8 T-cell responses, indicating that nodules are not required for aluminum's adjuvant effect. Moreover, the ability of aluminum salts to retain antigen in the body, the well-known depot effect, was unaffected by the absence of nodules. We conclude that aluminum adjuvants form fibrin-dependent nodules in vivo, that these nodules have properties of extracellular traps, and the nodules are not required for aluminum salts to act as adjuvants.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Aluminum/administration & dosage , Adjuvants, Immunologic/pharmacology , Aluminum/pharmacology , Animals , Fibrin/metabolism , Fibrinogen/metabolism , Histones/metabolism , Mice , Mice, Knockout , Salts , T-Lymphocytes/immunology , Thrombin/metabolism , Vaccination
14.
J Immunol ; 183(7): 4403-14, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19734227

ABSTRACT

To understand more about how the body recognizes alum we characterized the early innate and adaptive responses in mice injected with the adjuvant. Within hours of exposure, alum induces a type 2 innate response characterized by an influx of eosinophils, monocytes, neutrophils, DCs, NK cells and NKT cells. In addition, at least 13 cytokines and chemokines are produced within 4 h of injection including IL-1beta and IL-5. Optimal production of some of these, including IL-1beta, depends upon both macrophages and mast cells, whereas production of others, such as IL-5, depends on mast cells only, suggesting that both of these cell types can detect alum. Alum induces eosinophil accumulation partly through the production of mast cell derived IL-5 and histamine. Alum greatly enhances priming of endogenous CD4 and CD8 T cells independently of mast cells, macrophages, and of eosinophils. In addition, Ab levels and Th2 bias was similar in the absence of these cells. We found that the inflammation induced by alum was unchanged in caspase-1-deficient mice, which cannot produce IL-1beta. Furthermore, endogenous CD4 and CD8 T cell responses, Ab responses and the Th2 bias were also not impacted by the absence of caspase-1 or NLRP3. These data suggest that activation of the inflammasome and the type 2 innate response orchestrated by macrophages and mast cells in vivo are not required for adjuvant effect of alum on endogenous T and B cell responses.


Subject(s)
Adjuvants, Immunologic/pharmacology , Alum Compounds/pharmacology , Immunity, Cellular/drug effects , Immunity, Innate/drug effects , Inflammation Mediators/pharmacology , Macrophages, Peritoneal/immunology , Mast Cells/immunology , Adjuvants, Immunologic/administration & dosage , Alum Compounds/administration & dosage , Amino Acid Sequence , Animals , Biosensing Techniques , Carrier Proteins/physiology , Caspase 1/physiology , Cell Movement/drug effects , Cell Movement/immunology , Inflammation Mediators/administration & dosage , Inflammation Mediators/classification , Injections, Intraperitoneal , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mast Cells/drug effects , Mast Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , NLR Family, Pyrin Domain-Containing 3 Protein
15.
J Exp Med ; 206(3): 515-23, 2009 Mar 16.
Article in English | MEDLINE | ID: mdl-19255146

ABSTRACT

Natural killer (NK) cells are crucial in resistance to certain viral infections, but the mechanisms used to recognize infected cells remain largely unknown. Here, we show that the activating Ly49P receptor recognizes cells infected with mouse cytomegalovirus (MCMV) by a process that requires the presence of H2-D(k) and the MCMV m04 protein. Using H2 chimeras between H2-D(b) and -D(k), we demonstrate that the H2-D(k) peptide-binding platform is required for Ly49P recognition. We identified m04 as a viral component necessary for recognition using a panel of MCMV-deletion mutant viruses and complementation of m04-deletion mutant (Deltam04) virus infection. MA/My mice, which express Ly49P and H2-D(k), are resistant to MCMV; however, infection with Deltam04 MCMV abrogates resistance. Depletion of NK cells in MA/My mice abrogates their resistance to wild-type MCMV infection, but does not significantly affect viral titers in mice infected with Deltam04 virus, implicating NK cells in host protection through m04-dependent recognition. These findings reveal a novel mechanism of major histocompatibility complex class I-restricted recognition of virally infected cells by an activating NK cell receptor.


Subject(s)
Antiviral Agents/immunology , Carrier Proteins/immunology , Glycoproteins/immunology , Histocompatibility Antigens Class I/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Muromegalovirus/immunology , Receptors, Immunologic/immunology , Viral Proteins/immunology , Animals , Biological Assay , Herpesviridae Infections/immunology , Histocompatibility Antigens Class I/chemistry , Lymphocyte Activation , Mice , Mutation/genetics , NIH 3T3 Cells , Protein Structure, Tertiary
16.
Nat Rev Immunol ; 9(4): 287-93, 2009 04.
Article in English | MEDLINE | ID: mdl-19247370

ABSTRACT

The efficacy of vaccines depends on the presence of an adjuvant in conjunction with the antigen. Of these adjuvants, the ones that contain aluminium, which were first discovered empirically in 1926, are currently the most widely used. However, a detailed understanding of their mechanism of action has only started to be revealed. In this Timeline article, we briefly describe the initial discovery of aluminium adjuvants and discuss historically important advances. We also summarize recent progress in the field and discuss their implications and the remaining questions on how these adjuvants work.


Subject(s)
Adjuvants, Immunologic , Aluminum/immunology , Adsorption/immunology , Aluminum Compounds/chemistry , Aluminum Compounds/immunology , Aluminum Compounds/pharmacology , Animals , Antibody Formation/drug effects , Antibody Formation/immunology , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antigens/chemistry , Antigens/immunology , Apoptosis Regulatory Proteins , CARD Signaling Adaptor Proteins , Carrier Proteins/physiology , Caspase 1/physiology , Cytoskeletal Proteins/physiology , Humans , Immunity, Cellular/drug effects , Immunity, Cellular/immunology , Immunity, Innate/drug effects , Immunity, Innate/immunology , Mice , Models, Immunological , NLR Family, Pyrin Domain-Containing 3 Protein
17.
J Immunol ; 181(2): 1128-34, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18606665

ABSTRACT

The dynamics of mouse cytomegalovirus (MCMV)-specific CD4 T cell responses and the mechanisms by which these cells contribute to viral control are not well understood, mainly due to lack of appropriate tools to characterize MCMV-specific CD4 T cells. We therefore generated MCMV-specific CD4 T cell hybridomas, then used an MCMV expression library and overlapping peptides to identify CD4 T cell epitopes. We used these novel tools to study the long-term kinetics and organ distribution of MCMV-specific CD4 T cells in comparison to MCMV-specific CD8 T cell responses. We demonstrate that the overall MCMV-specific CD4 T cell response stabilizes during the latent stage, which stands in contrast to subpopulations of MCMV-specific CD8 T cells and HCMV-specific CD4 T cells which accumulate over the course of CMV latency. Furthermore, MCMV-specific CD4 T cells displayed a Th1 phenotype, secreting high levels of IFN-gamma and TNF-alpha and to some extent IL-2, cytokines which are involved in protection from CMV disease.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Epitopes, T-Lymphocyte/immunology , Herpesviridae Infections/immunology , Muromegalovirus/immunology , Virus Latency , Acute Disease , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line , Cytokines/immunology , Epitopes, T-Lymphocyte/metabolism , Herpesviridae Infections/virology , Humans , Mice , Mice, Inbred C57BL , Peptides/immunology , Peptides/metabolism
18.
J Virol ; 82(12): 5781-96, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18367531

ABSTRACT

Cytomegalovirus (CMV) infection continues to be a complication in recipients of hematopoietic stem cell transplantation (HSCT). Preexisting donor immunity is recognized as a favorable prognostic factor for the reconstitution of protective antiviral immunity mediated primarily by CD8 T cells. Furthermore, adoptive transfer of CMV-specific memory CD8 T (CD8-T(M)) cells is a therapeutic option for preventing CMV disease in HSCT recipients. Given the different CMV infection histories of donor and recipient, a problem may arise from an antigenic mismatch between the CMV variant that has primed donor immunity and the CMV variant acquired by the recipient. Here, we have used the BALB/c mouse model of CMV infection in the immunocompromised host to evaluate the importance of donor-recipient CMV matching in immundominant epitopes (IDEs). For this, we generated the murine CMV (mCMV) recombinant virus mCMV-DeltaIDE, in which the two memory repertoire IDEs, the IE1-derived peptide 168-YPHFMPTNL-176 presented by the major histocompatibility complex class I (MHC-I) molecule L(d) and the m164-derived peptide 257-AGPPRYSRI-265 presented by the MHC-I molecule D(d), are both functionally deleted. Upon adoptive transfer, polyclonal donor CD8-T(M) cells primed by mCMV-DeltaIDE and the corresponding revertant virus mCMV-revDeltaIDE controlled infection of immunocompromised recipients with comparable efficacy and regardless of whether or not IDEs were presented in the recipients. Importantly, CD8-T(M) cells primed under conditions of immunodomination by IDEs protected recipients in which IDEs were absent. This shows that protection does not depend on compensatory expansion of non-IDE-specific CD8-T(M) cells liberated from immunodomination by the deletion of IDEs. We conclude that protection is, rather, based on the collective antiviral potential of non-IDEs independent of the presence or absence of IDE-mediated immunodomination.


Subject(s)
Adoptive Transfer , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Herpesviridae Infections/immunology , Muromegalovirus/immunology , Animals , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Disease Models, Animal , Female , Fibroblasts/virology , Immunodominant Epitopes/genetics , Kinetics , Mice , Mice, Inbred BALB C , Viral Proteins/genetics , Viral Proteins/metabolism
19.
Immunity ; 27(5): 687-90, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18031690

ABSTRACT

In this Commentary, McKee et al. highlight the properties of extrinsic vaccine adjuvants that must be considered to achieve the most protective immune response, as occurs naturally with many intrinsic pathogen-derived adjuvants.


Subject(s)
Adjuvants, Immunologic/pharmacology , Immune System/physiology , Infections/immunology , Vaccines/immunology , Animals , Humans
20.
J Immunol ; 179(4): 2195-202, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17675479

ABSTRACT

The mechanisms that regulate CMV-specific T cell responses in vivo are poorly understood. During murine CMV infection of B6 mice, primary responses in the spleen are dominated by CD8 T cells reactive with antigenic epitopes in M45, M57, and m139 murine CMV gene products. However, during the later persistent phase of infection, CD8 T cell responses to epitopes in m139 and M38 viral gene products predominate. The basis for this shift in CD8 T populations is unknown. In this study, we demonstrate that OX40, a TNFR superfamily member, specifically regulates the accumulation of CD8 T cells reactive with the persistent-phase epitopes. Defective CD8 T cell responses in OX40(-/-) mice were replicated in MHC class II(-/-) mice implying that CD4 T cells in part controlled the differentiation of the CD8 T cell clones responsive to these epitopes during persistent infection. Furthermore, treatment of infected mice with an agonist OX40 Ab induced expansion of protective primary virus-specific CD8 T cells independent of CD4 T cell help, but CD4 T cells were crucial for anti-OX40 to promote CD8 T cells reactive to the persistent dominant epitopes. Collectively, these results indicate manipulation of OX40 may be useful in improving cellular immunotherapy regimes for treatment of persistent virus infections.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Herpesviridae Infections/immunology , Muromegalovirus/immunology , Receptors, OX40/immunology , Viral Proteins/immunology , Adoptive Transfer , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Proliferation , Herpesviridae Infections/therapy , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Mice , Mice, Knockout , Receptors, OX40/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...