Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
mBio ; 11(2)2020 03 03.
Article in English | MEDLINE | ID: mdl-32127460

ABSTRACT

Arthritogenic alphaviruses such as Ross River and Chikungunya viruses cause debilitating muscle and joint pain and pose significant challenges in the light of recent outbreaks. How host immune responses are orchestrated after alphaviral infections and lead to musculoskeletal inflammation remains poorly understood. Here, we show that myositis induced by Ross River virus (RRV) infection is driven by CD11bhi Ly6Chi inflammatory monocytes and followed by the establishment of a CD11bhi Ly6Clo CX3CR1+ macrophage population in the muscle upon recovery. Selective modulation of CD11bhi Ly6Chi monocyte migration to infected muscle using immune-modifying microparticles (IMP) reduced disease score, tissue damage, and inflammation and promoted the accumulation of CX3CR1+ macrophages, enhancing recovery and resolution. Here, we detail the role of immune pathology, describing a poorly characterized muscle macrophage subset as part of the dynamics of alphavirus-induced myositis and tissue recovery and identify IMP as an effective immunomodulatory approach. Given the lack of specific treatments available for alphavirus-induced pathologies, this study highlights a therapeutic potential for simple immune modulation by IMP in infected individuals in the event of large alphavirus outbreaks.IMPORTANCE Arthritogenic alphaviruses cause debilitating inflammatory disease, and current therapies are restricted to palliative approaches. Here, we show that following monocyte-driven muscle inflammation, tissue recovery is associated with the accumulation of CX3CR1+ macrophages in the muscle. Modulating inflammatory monocyte infiltration using immune-modifying microparticles (IMP) reduced tissue damage and inflammation and enhanced the formation of tissue repair-associated CX3CR1+ macrophages in the muscle. This shows that modulating key effectors of viral inflammation using microparticles can alter the outcome of disease by facilitating the accumulation of macrophage subsets associated with tissue repair.


Subject(s)
Alphavirus Infections/metabolism , Alphavirus Infections/virology , CX3C Chemokine Receptor 1/genetics , Monocytes/metabolism , Myositis/etiology , Myositis/metabolism , Wound Healing , Alphavirus Infections/pathology , Animals , Biomarkers , Biopsy , CX3C Chemokine Receptor 1/metabolism , Disease Models, Animal , Gene Expression Profiling , Immunomodulation/genetics , Immunophenotyping , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , Mice , Mice, Transgenic , Monocytes/immunology , Monocytes/virology , Myositis/pathology
2.
J Virol ; 90(8): 4150-4159, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26865723

ABSTRACT

UNLABELLED: The alphaviral6kgene region encodes the two structural proteins 6K protein and, due to a ribosomal frameshift event, the transframe protein (TF). Here, we characterized the role of the6kproteins in the arthritogenic alphavirus Ross River virus (RRV) in infected cells and in mice, using a novel6kin-frame deletion mutant. Comprehensive microscopic analysis revealed that the6kproteins were predominantly localized at the endoplasmic reticulum of RRV-infected cells. RRV virions that lack the6kproteins 6K and TF [RRV-(Δ6K)] were more vulnerable to changes in pH, and the corresponding virus had increased sensitivity to a higher temperature. While the6kdeletion did not reduce RRV particle production in BHK-21 cells, it affected virion release from the host cell. Subsequentin vivostudies demonstrated that RRV-(Δ6K) caused a milder disease than wild-type virus, with viral titers being reduced in infected mice. Immunization of mice with RRV-(Δ6K) resulted in a reduced viral load and accelerated viral elimination upon secondary infection with wild-type RRV or another alphavirus, chikungunya virus (CHIKV). Our results show that the6kproteins may contribute to alphaviral disease manifestations and suggest that manipulation of the6kgene may be a potential strategy to facilitate viral vaccine development. IMPORTANCE: Arthritogenic alphaviruses, such as chikungunya virus (CHIKV) and Ross River virus (RRV), cause epidemics of debilitating rheumatic disease in areas where they are endemic and can emerge in new regions worldwide. RRV is of considerable medical significance in Australia, where it is the leading cause of arboviral disease. The mechanisms by which alphaviruses persist and cause disease in the host are ill defined. This paper describes the phenotypic properties of an RRV6kdeletion mutant. The absence of the6kgene reduced virion release from infected cells and also reduced the severity of disease and viral titers in infected mice. Immunization with the mutant virus protected mice against viremia not only upon exposure to RRV but also upon challenge with CHIKV. These findings could lead to the development of safer and more immunogenic alphavirus vectors for vaccine delivery.


Subject(s)
Alphavirus Infections/virology , Ross River virus/genetics , Ross River virus/immunology , Viral Structural Proteins/genetics , Alphavirus Infections/immunology , Alphavirus Infections/physiopathology , Animals , Cell Line , Cell Line, Tumor , Chikungunya virus/immunology , Chlorocebus aethiops , Cricetinae , Humans , Hydrogen-Ion Concentration , Mice , Mutation , Reading Frames , Ross River virus/pathogenicity , Sequence Deletion , Vero Cells , Viral Load , Viral Structural Proteins/analysis , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/immunology , Virus Replication
3.
J Virol ; 89(15): 8063-76, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26018160

ABSTRACT

UNLABELLED: Arthritogenic alphaviruses such as Ross River virus (RRV) and chikungunya virus (CHIKV) cause large-scale epidemics of severe musculoskeletal disease and have been progressively expanding their global distribution. Since its introduction in July 2014, CHIKV now circulates in the United States. The hallmark of alphavirus disease is crippling pain and inflammation of the joints, a similar immunopathology to rheumatoid arthritis. The use of glycans as novel therapeutics is an area of research that has increased in recent years. Here, we describe the promising therapeutic potential of the glycosaminoglycan (GAG)-like molecule pentosan polysulfate (PPS) to alleviate virus-induced arthritis. Mouse models of RRV and CHIKV disease were used to characterize the extent of cartilage damage in infection and investigate the potential of PPS to treat disease. This was assessed using histological analysis, real-time PCR, and fluorescence-activated cell sorting (FACS). Alphaviral infection resulted in cartilage destruction, the severity of which was alleviated by PPS therapy during RRV and CHIKV clinical disease. The reduction in cartilage damage corresponded with a significant reduction in immune infiltrates. Using multiplex bead arrays, PPS treatment was found to have significantly increased the anti-inflammatory cytokine interleukin-10 and reduced proinflammatory cytokines, typically correlated with disease severity. Furthermore, we reveal that the severe RRV-induced joint pathology, including thinning of articular cartilage and loss of proteoglycans in the cartilage matrix, was diminished with treatment. PPS is a promising new therapy for alphavirus-induced arthritis, acting to preserve the cartilage matrix, which is damaged during alphavirus infection. Overall, the data demonstrate the potential of glycotherapeutics as a new class of treatment for infectious arthritis. IMPORTANCE: The hallmark of alphavirus disease is crippling pain and joint arthritis, which often has an extended duration. In the past year, CHIKV has expanded into the Americas, with approximately 1 million cases reported to date, whereas RRV continues to circulate in the South Pacific. Currently, there is no licensed specific treatment for alphavirus disease, and the increasing spread of infection highlights an urgent need for therapeutic intervention strategies. Pentosan polysulfate (PPS) is a glycan derivative that is orally bioavailable, has few toxic side effects, and is currently licensed under the name Elmiron for the treatment of cystitis in the United States. Our findings show that RRV infection damages the articular cartilage, including a loss of proteoglycans within the joint. Furthermore, treatment with PPS reduced the severity of both RRV- and CHIKV-induced musculoskeletal disease, including a reduction in inflammation and joint swelling, suggesting that PPS is a promising candidate for drug repurposing for the treatment of alphavirus-induced arthritis.


Subject(s)
Cartilage/immunology , Chikungunya Fever/drug therapy , Chikungunya virus/physiology , Glycosaminoglycans/administration & dosage , Joint Diseases/drug therapy , Pentosan Sulfuric Polyester/administration & dosage , Animals , Cartilage/drug effects , Cartilage/virology , Chikungunya Fever/immunology , Chikungunya Fever/virology , Disease Models, Animal , Humans , Joint Diseases/immunology , Joint Diseases/virology , Mice , Mice, Inbred C57BL
4.
PLoS Pathog ; 11(2): e1004649, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25695775

ABSTRACT

The rising prevalence of arthritogenic alphavirus infections, including chikungunya virus (CHIKV) and Ross River virus (RRV), and the lack of antiviral treatments highlight the potential threat of a global alphavirus pandemic. The immune responses underlying alphavirus virulence remain enigmatic. We found that pentraxin 3 (PTX3) was highly expressed in CHIKV and RRV patients during acute disease. Overt expression of PTX3 in CHIKV patients was associated with increased viral load and disease severity. PTX3-deficient (PTX3(-/-)) mice acutely infected with RRV exhibited delayed disease progression and rapid recovery through diminished inflammatory responses and viral replication. Furthermore, binding of the N-terminal domain of PTX3 to RRV facilitated viral entry and replication. Thus, our study demonstrates the pivotal role of PTX3 in shaping alphavirus-triggered immunity and disease and provides new insights into alphavirus pathogenesis.


Subject(s)
Alphavirus Infections/immunology , C-Reactive Protein/immunology , Nerve Tissue Proteins/immunology , Serum Amyloid P-Component/immunology , Virus Replication/immunology , Animals , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Transcriptome , Transfection , Viral Load/immunology
5.
J Gen Virol ; 95(Pt 10): 2146-2154, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24934444

ABSTRACT

Alphaviruses including Barmah Forest virus (BFV) and Ross River virus (RRV) cause arthritis, arthralgia and myalgia in humans. The rheumatic symptoms in human BFV infection are very similar to those of RRV. Although RRV disease has been studied extensively, little is known about the pathogenesis of BFV infection. We sought to establish a mouse model for BFV to facilitate our understanding of BFV infectivity, tropism and pathogenesis, and to identify key pathological and immunological mechanisms of BFV infection that may distinguish between infections with BFV and RRV. Here, to the best of our knowledge, we report the first study assessing the virulence and replication of several BFV isolates in a mouse model. We infected newborn Swiss outbred mice with BFV and established that the BFV2193 prototype was the most virulent strain. BFV2193 infection resulted in the highest mortality among all BFV variant isolates, comparable to that of RRV. In comparison with RRV, C57BL/6 mice infected with BFV showed delayed onset, moderate disease scores and early recovery of the disease. BFV replicated poorly in muscle and did not cause the severe myositis seen in RRV-infected mice. The mRNAs for the inflammatory mediators TNF-α, IL-6, CCL2 and arginase-1 were highly upregulated in RRV- but not BFV-infected muscle. To our knowledge, this is the first report of a mouse model of BFV infection, which we have used to demonstrate differences between BFV and RRV infections and to further understand disease pathogenesis. With an increasing number of BFV cases occurring annually, a better understanding of the disease mechanisms is essential for future therapeutic development.


Subject(s)
Alphavirus Infections/pathology , Alphavirus Infections/virology , Alphavirus/physiology , Alphavirus/immunology , Alphavirus/pathogenicity , Alphavirus Infections/immunology , Animals , Animals, Newborn , Cytokines/biosynthesis , Disease Models, Animal , Female , Gene Expression Profiling , Mice , Mice, Inbred C57BL , Survival Analysis , Virulence , Virus Replication
6.
Proc Natl Acad Sci U S A ; 111(16): 6040-5, 2014 Apr 22.
Article in English | MEDLINE | ID: mdl-24733914

ABSTRACT

Arthritogenic alphaviruses including Ross River virus (RRV), Sindbis virus, and chikungunya virus cause worldwide outbreaks of musculoskeletal disease. The ability of alphaviruses to induce bone pathologies remains poorly defined. Here we show that primary human osteoblasts (hOBs) can be productively infected by RRV. RRV-infected hOBs produced high levels of inflammatory cytokine including IL-6. The RANKL/OPG ratio was disrupted in the synovial fluid of RRV patients, and this was accompanied by an increase in serum Tartrate-resistant acid phosphatase 5b (TRAP5b) levels. Infection of bone cells with RRV was validated using an established RRV murine model. In wild-type mice, infectious virus was detected in the femur, tibia, patella, and foot, together with reduced bone volume in the tibial epiphysis and vertebrae detected by microcomputed tomographic (µCT) analysis. The RANKL/OPG ratio was also disrupted in mice infected with RRV; both this effect and the bone loss were blocked by treatment with an IL-6 neutralizing antibody. Collectively, these findings provide previously unidentified evidence that alphavirus infection induces bone loss and that OBs are capable of producing proinflammatory mediators during alphavirus-induced arthralgia. The perturbed RANKL/OPG ratio in RRV-infected OBs may therefore contribute to bone loss in alphavirus infection.


Subject(s)
Alphavirus Infections/pathology , Alphavirus Infections/virology , Arthritis/virology , Bone Resorption/pathology , Bone Resorption/virology , Osteoblasts/pathology , Ross River virus/physiology , Acid Phosphatase/blood , Adult , Alphavirus Infections/blood , Animals , Antibodies, Neutralizing/pharmacology , Arthritis/blood , Arthritis/pathology , Bone Resorption/blood , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Bone and Bones/virology , Female , Growth Plate/drug effects , Growth Plate/pathology , Growth Plate/virology , Humans , Inflammation Mediators/metabolism , Interleukin-6/biosynthesis , Isoenzymes/blood , Male , Mice , Mice, Inbred C57BL , Neutralization Tests , Osteoblasts/drug effects , Osteoblasts/virology , Osteoclasts/drug effects , Osteoclasts/pathology , Osteoclasts/virology , Osteogenesis/drug effects , Osteoprotegerin/metabolism , Phenotype , RANK Ligand/metabolism , Ross River virus/drug effects , Synovial Fluid/metabolism , Tartrate-Resistant Acid Phosphatase , Virus Replication/drug effects , X-Ray Microtomography
7.
PLoS One ; 9(4): e95208, 2014.
Article in English | MEDLINE | ID: mdl-24743235

ABSTRACT

Unraveling the mechanisms of hematopoiesis regulated by multiple cytokines remains a challenge in hematology. IL-3 is an allergic cytokine with the multilineage potential, while CSF-1 is produced in the steady state with restricted lineage coverage. Here, we uncovered an instructive role of CSF-1 in IL-3-mediated hematopoiesis. CSF-1 significantly promoted IL-3-driven CD11c+ cell expansion and dampened basophil and mast cell generation from C57BL/6 bone marrow. Further studies indicated that the CSF-1/CSF-1R axis contributed significantly to IL-3-induced CD11c+ cell generation through enhancing c-Fos-associated monopoiesis. CD11c+ cells induced by IL-3 or IL-3/CSF-1 were competent in cellular maturation and endocytosis. Both IL-3 and IL-3/CSF-1 cells lacked classical dendritic cell appearance and resembled macrophages in morphology. Both populations produced a high level of IL-10, in addition to IL-1, IL-6 and TNFα, in response to LPS, and were relatively poor T cell stimulators. Collectively, these findings reveal a role for CSF-1 in mediating the IL-3 hematopoietic pathway through monopoiesis, which regulates expansion of CD11c+ macrophages.


Subject(s)
CD11c Antigen/immunology , Interleukin-10/immunology , Interleukin-3/immunology , Macrophage Colony-Stimulating Factor/immunology , Macrophages/immunology , Myelopoiesis/immunology , Animals , Interleukin-1/immunology , Interleukin-6/immunology , Macrophages/cytology , Mice , Mice, Inbred BALB C , Tumor Necrosis Factor-alpha/immunology
8.
PLoS One ; 8(8): e71146, 2013.
Article in English | MEDLINE | ID: mdl-23951095

ABSTRACT

Part of the Togaviridae family, alphaviruses, including chikungunya virus (CHIKV), Sindbis virus (SINV) and Ross River virus (RRV), are able to cause significant inflammatory pathologies ranging from arthritis to encephalitis. Following symptomatic infection with arthritis-associated alphaviruses, patients often experience severe joint pain, affecting distal and small joints, which can last six months or longer. Recently, methotrexate (MTX), a disease modifying anti-rheumatic drug (DMARD), was used to treat patients experiencing chronic rheumatic symptoms following infection with CHIKV. Here, the effect of MTX on Ross River virus disease (RRVD) in mice was examined to better understand its therapeutic potential for alphaviral-induced musculoskeletal disease and to further our knowledge of the development of alphaviral pathologies. Using a mouse model, we analyzed the effect of MTX on RRVD. RRV disease pathogenesis in response to MTX treatment was determined by measuring levels of proinflammatory factors, cellular infiltrates, viral titer and histological analysis of infected tissues. RRV-infected mice receiving MTX treatment rapidly developed musculoskeletal disease, which correlated with a significant influx of inflammatory cell infiltrates into the skeletal muscle tissue. Although no difference was observed in the level of proinflammatory cytokines and chemokines, the viral load increased at early time points post infection in the serum and quadriceps of MTX treated mice, possibly contributing to disease pathogenesis. Results suggest that MTX treatment of acute RRVD in mice provides no therapeutic benefit and underline the importance of inflammatory monocytes in alphaviral induced arthritides.


Subject(s)
Alphavirus Infections/complications , Alphavirus Infections/immunology , Antirheumatic Agents/adverse effects , Inflammation/etiology , Methotrexate/adverse effects , Monocytes/immunology , Ross River virus , Alphavirus Infections/drug therapy , Animals , Antirheumatic Agents/therapeutic use , Arthritis/etiology , CD11b Antigen/metabolism , Disease Models, Animal , Inflammation Mediators/metabolism , Methotrexate/therapeutic use , Mice , Monocytes/metabolism , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology
9.
Arthritis Rheum ; 65(10): 2724-36, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23896945

ABSTRACT

OBJECTIVE: Arthrogenic alphaviruses such as Ross River virus (RRV) and chikungunya virus (CHIKV) circulate worldwide. This virus class causes debilitating illnesses that are characterized by arthritis, arthralgia, and myalgia. In previous studies, we identified macrophage migration inhibitory factor (MIF) as a critical inflammatory factor in the pathogenesis of alphaviral diseases. The present study was undertaken to characterize the role of CD74, a cell surface receptor of MIF, in both RRV- and CHIKV-induced alphavirus arthritides. METHODS: Mouse models of RRV and CHIKV infection were used to investigate the immunopathogenesis of arthritic alphavirus infection. The role of CD74 was assessed using histologic analysis, real-time polymerase chain reaction, flow cytometry, and plaque assay. RESULTS: In comparison to wild-type mice, CD74-/- mice developed only mild clinical features and had low levels of tissue damage. Leukocyte infiltration, characterized predominantly by inflammatory monocytes and natural killer cells, was substantially reduced in the infected tissue of CD74-/- mice, but production of proinflammatory cytokines and chemokines was not decreased. CD74 deficiency was associated with increased monocyte apoptosis, but had no effect on monocyte migratory capacity. Consistent with these findings, alphaviral infection resulted in a dose-dependent up-regulation of CD74 expression in human peripheral blood mononuclear cells, and serum MIF levels were significantly elevated in patients with RRV or CHIKV infection. CONCLUSION: CD74 appears to regulate immune responses to alphaviral infection through its effects on cellular recruitment and survival. These findings suggest that both MIF and CD74 play a critical role in mediating alphaviral disease, and blocking these factors with novel therapeutic agents could substantially ameliorate the pathologic manifestations.


Subject(s)
Alphavirus Infections/complications , Antigens, Differentiation, B-Lymphocyte/physiology , Arthritis, Infectious/etiology , Arthritis, Infectious/physiopathology , Histocompatibility Antigens Class II/physiology , Myositis/physiopathology , Myositis/virology , Receptors, Immunologic/physiology , Alphavirus Infections/pathology , Animals , Antigens, Differentiation, B-Lymphocyte/genetics , Apoptosis/physiology , Arthritis, Infectious/pathology , Cells, Cultured , Chemokines/metabolism , Chikungunya virus/physiology , Cytokines/metabolism , Disease Models, Animal , Female , Histocompatibility Antigens Class II/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/pathology , Myositis/pathology , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Ross River virus/physiology , Severity of Illness Index
10.
J Drug Deliv ; 2013: 516749, 2013.
Article in English | MEDLINE | ID: mdl-23781340

ABSTRACT

Phenotypic maturation and T cell stimulation are two functional attributes of DCs critical for immune induction. The combination of antigens, including those from cancer, with Toll-like receptor (TLR) ligands induces far superior cellular immune responses compared to antigen alone. In this study, IFN-gamma treatment of bone marrow-derived DC, followed by incubation with the TLR2, TLR4, or TLR9 agonists, enhanced DC activation compared to TLR ligation alone. Most notably, the upregulation of CD40 with LPS stimulation and CD86 with CpG stimulation was observed in in vitro cultures. Similarly, IFN-gamma coinjected with TLR ligands was able to promote DC activation in vivo, with DCs migrating from the site of immunization to the popliteal lymph nodes demonstrating increased expression of CD80 and CD86. The heightened DC activation translated to a drastic increase in T cell stimulatory capacity in both antigen independent and antigen dependent fashions. This is the first time that IFN-gamma has been shown to have a combined effect with TLR ligation to enhance DC activation and function. The results demonstrate the novel use of IFN-gamma together with TLR agonists to enhance antigen-specific T cell responses, for applications in the development of enhanced vaccines and drug targets against diseases including cancer.

11.
Pharmacol Ther ; 137(2): 266-82, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23103333

ABSTRACT

Dengue virus (DV) is the most widespread arbovirus, being endemic in over 100 countries, and is estimated to cause 50 million infections annually. Viral factors, such as the genetic composition of the virus strain can play a role in determining the virus virulence and subsequent clinical disease severity. Virus vector competence plays an integral role in virus transmission and is a critical factor in determining the severity and impact of DV outbreaks. Host genetic variations in immune-related genes, including the human leukocyte antigen, have also been shown to correlate with clinical disease and thus may play a role in regulating disease severity. The host's immune system, however, appears to be the primary factor in DV pathogenesis with the delicate interplay of innate and acquired immunity playing a crucial role. Although current research of DV pathogenesis has been limited by the lack of an appropriate animal model, the development of DV therapeutics has been a primary focus of research groups around the world. In the past decade advances in both the development of vaccines and anti-virals have increased in dramatically. This review summarises the current understanding of viral, vector and host factors which contribute to dengue virus pathogenesis and how this knowledge is critically important in the development of pharmaceutical interventions.


Subject(s)
Dengue Virus/pathogenicity , Dengue/etiology , Genetic Predisposition to Disease , Host-Derived Cellular Factors , Insect Vectors , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Dengue/immunology , Dengue/prevention & control , Dengue/virology , Dengue Virus/genetics , Dengue Virus/metabolism , Host-Derived Cellular Factors/genetics , Host-Derived Cellular Factors/immunology , Humans , Viral Proteins/genetics , Viral Proteins/metabolism , Viral Vaccines/immunology , Viral Vaccines/pharmacology
12.
J Immunol ; 185(6): 3158-66, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20709950

ABSTRACT

The cooperative nature of tetraspanin-tetraspanin interactions in membrane organization suggests functional overlap is likely to be important in tetraspanin biology. Previous functional studies of the tetraspanins CD37 and Tssc6 in the immune system found that both CD37 and Tssc6 regulate T cell proliferative responses in vitro. CD37(-/-) mice also displayed a hyper-stimulatory dendritic cell phenotype and dysregulated humoral responses. In this study, we characterize "double knockout" mice (CD37(-/-)Tssc6(-/-)) generated to investigate functional overlap between these tetraspanins. Strong evidence for a cooperative role for these two proteins was identified in cellular immunity, where both in vitro T cell proliferative responses and dendritic cell stimulation capacity are significantly exaggerated in CD37(-/-)Tssc6(-/-) mice when compared with single knockout counterparts. Despite these exaggerated cellular responses in vitro, CD37(-/-)Tssc6(-/-) mice are not more susceptible to autoimmune induction. However, in vivo responses to pathogens appear poor in CD37(-/-)Tssc6(-/-) mice, which showed a reduced ability to produce influenza-specific T cells and displayed a rapid onset hyper-parasitemia when infected with Plasmodium yoelii. Therefore, in the absence of both CD37 and Tssc6, immune function is further altered when compared with CD37(-/-) or Tssc6(-/-) mice, demonstrating a complementary role for these two molecules in cellular immunity.


Subject(s)
Antigens, CD/physiology , Antigens, Neoplasm/physiology , Dendritic Cells/immunology , Membrane Proteins/physiology , T-Lymphocyte Subsets/immunology , Amino Acid Sequence , Animals , Antigens, CD/genetics , Antigens, Neoplasm/genetics , Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Cells, Cultured , Dendritic Cells/metabolism , Dendritic Cells/virology , Humans , Immunophenotyping , Influenza, Human/genetics , Influenza, Human/immunology , Influenza, Human/pathology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Malaria/genetics , Malaria/immunology , Malaria/pathology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/virology , Tetraspanins
13.
Expert Rev Vaccines ; 9(6): 595-600, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20518715

ABSTRACT

A successful vaccine for immunotherapy, particularly for solid tumors or viral infections, requires a suitable target antigen and the production of a cytotoxic T-cell response. In addition, CD4 T cells play an important role in cellular immunity. Here, we briefly discuss methods by which T cells are measured in vitro after vaccination.


Subject(s)
Cancer Vaccines/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Hematopoietic Stem Cells/immunology , Humans , Neoplasms/immunology , Ovalbumin/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccination
14.
Biochim Biophys Acta ; 1798(12): 2286-95, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20478265

ABSTRACT

Cell penetrating peptides (CPP) represent a novel approach to facilitate cytoplasmic delivery of macromolecules. The DNA binding domain of Drosophila Antennapedia contains 60 amino acids and consists of 3 α-helices, with internalizing activity mapped to a 16-amino acid peptide penetratin (Antp) within the third α-helix. Here, we report on the use of penetratin to deliver a multiple antigen peptide (MAP) incorporating the immunodominant CD8 epitope of ovalbumin, SIINFEKL (MAPOVACD8). We demonstrate that penetratin linked to the MAPOVACD8 construct either by a disulfide (SS) or thioether (SC) linkage promotes the uptake, cross presentation and subsequent in vivo proliferation and generation of OVACD8 (SIINFEKL)-specific T cells. The MAPOVACD8 construct without penetratin is not presented by MHC class I molecules nor does it generate an in vivo IFN-γ response in C57BL/6 mice. Moreover, we clearly define the uptake and intracellular processing pathways of AntpMAPOVACD8 SS and SC revealing the majority of AntpMAPOVACD8 is taken up by DC via an endocytic, proteasome and tapasin independent mechanism. We also show that the uptake mechanism of AntpMAPOVACD8 is dose dependent and uptake or intracellular processing is not altered by the type of chemical linkage.


Subject(s)
Carrier Proteins , Cell-Penetrating Peptides , Epitopes , Ovalbumin , T-Lymphocytes/immunology , Animals , Antigen Presentation/drug effects , Antigen Presentation/immunology , Carrier Proteins/chemistry , Carrier Proteins/immunology , Carrier Proteins/pharmacology , Cell Proliferation/drug effects , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/immunology , Cell-Penetrating Peptides/pharmacology , Dendritic Cells/immunology , Dose-Response Relationship, Drug , Dose-Response Relationship, Immunologic , Drosophila , Endocytosis/drug effects , Endocytosis/immunology , Epitopes/chemistry , Epitopes/immunology , Epitopes/pharmacology , Histocompatibility Antigens Class I/immunology , Interferon-gamma/immunology , Membrane Transport Proteins/immunology , Mice , Ovalbumin/chemistry , Ovalbumin/immunology , Ovalbumin/pharmacology , Proteasome Endopeptidase Complex/immunology , Protein Structure, Secondary
15.
J Immunol ; 184(6): 2863-72, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20176741

ABSTRACT

Reactive oxygen species (ROS) have been implicated in various physiological activities. However, their role in dendritic cell (DC) activation and generation has not been investigated. Using the bone marrow-derived GM-CSF-induced ex vivo DC model, we characterize how induction of ROS correlates with inflammatory DC functionality and expansion. We describe that the functionality of GM-CSF-induced DCs is distinct in two developmental stages. Whereas division of DC-committed hematopoietic progenitor cells (HPCs) neared completion by day 6, the level of ROS soared after day 4. Day 3 ROS(lo) DCs were highly responsive to TLR stimuli such as LPS and zymosan by rapid upregulation of CD80, CD86, and MHC class II, in contrast to the low response of day 6 ROS(hi) DCs. ROS(hi) DCs could not initiate and sustain a significant level of NF-kappaB phosphorylation in response to LPS and zymosan, although demonstrating hyperactivation of p38 MAPK by LPS, in a fashion disparate to ROS(lo) DCs. ROS(lo) DCs stimulated a higher level of allogeneic and OVA-specific T cell proliferative responses, although ROS(hi) DCs were much more proficient in processing OVA. In response to pathogenic stimuli, ROS(hi) DCs also demonstrated rapid cellular adhesion and H(2)O(2) release, indicating their role in immediate microbial targeting. Moreover, HPC expansion and DC generation were dependent on the surge of ROS in an NADPH oxidase-independent manner. These findings point to the potential role of cellular ROS in mediating functionality and development of DCs from HPCs during inflammation.


Subject(s)
Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Differentiation/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Inflammation Mediators/physiology , Reactive Oxygen Species/metabolism , Amino Acid Sequence , Animals , Bone Marrow Cells/immunology , Cells, Cultured , Dendritic Cells/immunology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Up-Regulation/immunology
16.
Eur J Immunol ; 39(1): 50-5, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19089816

ABSTRACT

A major question in immunology is how DC can display limited amounts of individual peptide-MHC complexes and still induce cross-linking of T-cell receptors to initiate cellular responses. One suggested mechanism is that MHC exists at the cell surface in high avidity multimers, and tetraspanin proteins, known to laterally associate with both MHC classes I and II, promote MHC multimerisation. To validate this theory, we tested the ability of DC deficient in either one of two typical tetraspanin molecules: CD37 or CD151 to present peptide to Ag-specific T cells. Surprisingly, although they exhibited no developmental or maturation defects, DC lacking either CD37 or CD151 expression were hyper-stimulatory to T cells. We demonstrate that CD37 and CD151 control DC-mediated T-cell activation by two different mechanisms: CD151 regulates co-stimulation whereas CD37 regulates peptide/MHC presentation. The implications of these results on the model suggesting that tetraspanins promote MHC multimerisation are discussed.


Subject(s)
Antigen Presentation , Antigens, CD/immunology , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Glycoproteins/immunology , Lymphocyte Activation , Animals , Antigen Presentation/genetics , Antigens, CD/genetics , Antigens, Neoplasm/genetics , Glycoproteins/genetics , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Tetraspanin 24 , Tetraspanins
17.
Biomaterials ; 30(7): 1389-400, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19058846

ABSTRACT

Receptor mediated gene delivery is an attractive non-viral method for targeting genetic material to specific cell types. We have previously utilized oxidized (OMPLL) and reduced mannan poly-L-lysine (RMPLL) to target DNA vaccines to antigen presenting cells and demonstrated that it could induce far stronger immune responses in mice compared to naked DNA immunization. In this study, we describe the immune enhancing attributes of mannan-PLL mediated DNA vaccination at the molecular level. Several attributes observed in similar gene delivery conjugates, such as entry via the endocytic pathway, low toxicity, protection from nucleases and compaction of particle size, were also evident here. In addition, OMPLL and RMPLL conjugates had profound effects on the antigen presentation functions of dendritic cells and macrophages, through the stimulation of cytokine production and maturation of dendritic cells. Interestingly, we demonstrate that OMPLL-DNA and RMPLL-DNA are able to mediate dendritic cell activation via toll-like receptor 2 as opposed to mannan alone which mediates via toll-like receptor 4. Overall, this report leads to greater understanding of how oxidized and reduced mannan mediated gene delivery could augment immune responses to DNA vaccination and provide insights into ways of further improving its immunogenicity.


Subject(s)
Drug Carriers , Gene Transfer Techniques , Mannans , Vaccines, DNA , Animals , B7-2 Antigen/immunology , Biocompatible Materials/chemistry , Biocompatible Materials/metabolism , Cytokines/immunology , Dendritic Cells/immunology , Dendritic Cells/physiology , Drug Carriers/chemistry , Humans , Mannans/chemistry , Mannans/immunology , Materials Testing , Mice , Mice, Inbred Strains , Mice, Knockout , Particle Size , Vaccines, DNA/chemistry , Vaccines, DNA/immunology
18.
Expert Rev Vaccines ; 7(7): 1005-18, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18767950

ABSTRACT

C-type lectin receptors (CLRs) are a class of pathogen-recognition receptors that are actively investigated in the field of vaccine delivery. Many of their properties have functions linked to the immune system. These receptors are expressed abundantly on antigen-presenting cells and are considered to be the sentinels of immune surveillance owing to their endocytic nature and the ability to recognize a diverse range of pathogens through recognition of pathogen-associated molecular patterns. CLRs are also involved in the processes of antigen presentation mediated through the induction of dendritic cell maturation and cytokine production. These properties engender CLRs to be ideal for vaccine targeting. Conversely, CLRs also function to recognize glycosylated self-antigens to induce homeostatic control and tolerance. In this review, we will describe the various preclinical/clinical vaccination strategies to target antigens and plasmid DNA to this diverse class of receptors.


Subject(s)
Lectins, C-Type/metabolism , Vaccination/methods , Vaccines, DNA/metabolism , Vaccines, Subunit/metabolism , Humans , Lectins, C-Type/immunology , Vaccines, DNA/immunology , Vaccines, Subunit/immunology
19.
J Immunol ; 181(4): 2455-64, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18684936

ABSTRACT

The evidence that dendritic cell (DC) subsets produce differential cytokines in response to specific TLR stimulation is robust. However, the role of TLR stimulation in Ag presentation and phenotypic maturation among DC subsets is not clear. Through the adjuvanticity of a novel mannosylated Ag, mannosylated dendrimer OVA (MDO), as a pathogen-associated molecular pattern Ag, we characterized the functionality of GM-CSF/IL-4-cultured bone marrow DC and Flt3 ligand (Flt3-L) DC subsets by Ag presentation and maturation assays. It was demonstrated that both bone marrow DCs and Flt3-L DCs bound, processed, and presented MDO effectively. However, while Flt3-L CD24(high) (conventional CD8(+) equivalent) and CD11b(high) (CD8(-) equivalent) DCs were adept at MDO processing by MHC class I and II pathways, respectively, CD45RA(+) plasmacytoid DCs presented MDO poorly to T cells. Successful MDO presentation was largely dependent on competent TLR4 for Ag localization and morphological/phenotypic maturation of DC subsets, despite the indirect interaction of MDO with TLR4. Furthermore, Toll/IL-1 receptor-domain-containing adaptor-inducing IFN-beta, but not MyD88, as a TLR4 signaling modulator was indispensable for MDO-induced DC maturation and Ag presentation. Taken together, our findings suggest that DC subsets differentially respond to a pathogen-associated molecular pattern-associated Ag depending on the intrinsic programming and TLRs expressed. Optimal functionality of DC subsets in Ag presentation necessitates concomitant TLR signaling critical for efficient Ag localization and processing.


Subject(s)
Adjuvants, Immunologic/physiology , Antigens/metabolism , Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Mannose/metabolism , Ovalbumin/immunology , Toll-Like Receptor 4/physiology , Amino Acid Sequence , Animals , Antigen Presentation/immunology , Antigens/immunology , Bone Marrow Cells/classification , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Communication/genetics , Cell Communication/immunology , Cell Differentiation/genetics , Cells, Cultured , Dendritic Cells/classification , Dendritic Cells/metabolism , Immunophenotyping , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Ovalbumin/chemical synthesis , Ovalbumin/metabolism , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
20.
Vaccine ; 26(31): 3827-34, 2008 Jul 23.
Article in English | MEDLINE | ID: mdl-18550230

ABSTRACT

DNA immunization is an attractive form of vaccination, which has shown promising results only in small animal models. There is a need to develop efficient gene delivery systems. We previously demonstrated that oxidized (OM) and reduced mannan (RM) complexed to ovalbumin DNA via poly-l-lysine (PLL), were able to generate potent immune responses in mice. Herein, we further investigated the suitability of OMPLL and RMPLL as carriers for mucin 1 (MUC1) DNA vaccination for cancer immunotherapy. Studies presented here showed that immune responses in C57BL/6 mice induced by OMPLL-MUC1 DNA and RMPLL-MUC1 DNA immunization were more immunogenic compared to MUC1 DNA alone. Moreover, tumor protection was evident at a dose as low as 0.5 microg. In addition, strong T cell responses were induced in HLA-A2 transgenic and human MUC1 transgenic mice. These results demonstrate the potential of OM and RM as efficient non-viral gene delivery carriers for DNA vaccines for use in cancer immunotherapy.


Subject(s)
Cancer Vaccines/immunology , Mannans/immunology , Mannans/metabolism , Mucin-1/immunology , Vaccines, DNA/immunology , Animals , Cytokines/metabolism , HLA-A2 Antigen/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mucin-1/genetics , Neoplasms/pathology , Neoplasms/prevention & control , Oxidation-Reduction , Survival Analysis , Vaccines, DNA/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...