Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Melanoma Res ; 29(4): 349-357, 2019 08.
Article in English | MEDLINE | ID: mdl-30615012

ABSTRACT

The cancer/testis antigen (CTA) family is a group of antigens whose expression is restricted to male germline cells of the testis and various malignancies. This expression pattern makes this group of antigens potential targets for immunotherapy. The aim of this study was to create an overview of CTA expressed by melanoma cells at mRNA and protein level. A systematic literature search was performed in Medline (PubMed) and Embase from inception up to and including February 2018. Studies were screened for eligibility by two independent reviewers. A total of 65 full-text articles were included in the final analysis. A total of 48 CTA have been studied in melanoma. Various CTA show different expression rates in primary and metastatic tumours. Of the 48 CTA, the most studied were MAGE-A3, MAGE-A1, NY-ESO-1, MAGE-A4, SSX2, MAGE-A2, MAGE-C1/CT7, SSX1, MAGE-C2/CT10 and MAGE-A12. On average, MAGE-A3 mRNA is present in 36% of primary tumours, whereas metastatic tumours have an expression rate of 55-81%. The same applies to the protein expression rate of MAGE-A3 in primary tumours, which is reported to be at 15-37%, whereas metastatic tumours have a higher expression rate of 25-70%. This trend of increased expression in metastases compared with primary tumours is observed with MAGE-A1, MAGE-A2, MAGE-A4, MAGE-A12 and NY-ESO-1. Many CTA are expressed on melanoma. This review provides an overview of the expression frequency of CTAs in melanoma and may aid in identifying CTA as the therapeutic target for immunotherapy.


Subject(s)
Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Melanoma/genetics , Skin Neoplasms/genetics , Testis/metabolism , Humans , Male , Melanoma/pathology , Skin Neoplasms/pathology , Melanoma, Cutaneous Malignant
2.
Infect Immun ; 83(10): 3881-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26195548

ABSTRACT

Foxp3(+) regulatory T (Treg) cells play a key role in suppression of immune responses during parasitic helminth infection, both by controlling damaging immunopathology and by inhibiting protective immunity. During the patent phase of Schistosoma mansoni infection, Foxp3(+) Treg cells are activated and suppress egg-elicited Th2 responses, but little is known of their induction and role during the early prepatent larval stage of infection. We quantified Foxp3(+) Treg cell responses during the first 3 weeks of murine S. mansoni infection in C57BL/6 mice, a time when larval parasites migrate from the skin and transit the lungs en route to the hepatic and mesenteric vasculature. In contrast to other helminth infections, S. mansoni did not elicit a Foxp3(+) Treg cell response during this early phase of infection. We found that the numbers and proportions of Foxp3(+) Treg cells remained unchanged in the lungs, draining lymph nodes, and spleens of infected mice. There was no increase in the activation status of Foxp3(+) Treg cells upon infection as assessed by their expression of CD25, Foxp3, and Helios. Furthermore, infection failed to induce Foxp3(+) Treg cells to produce the suppressive cytokine interleukin 10 (IL-10). Instead, only CD4(+) Foxp3(-) IL-4(+) Th2 cells showed increased IL-10 production upon infection. These data indicate that Foxp3(+) Treg cells do not play a prominent role in regulating immunity to S. mansoni larvae and that the character of the initial immune response invoked by S. mansoni parasites contrasts with the responses to other parasitic helminth infections that promote rapid Foxp3(+) Treg cell responses.


Subject(s)
Forkhead Transcription Factors/immunology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Movement , Female , Forkhead Transcription Factors/genetics , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Larva/immunology , Larva/physiology , Lung/immunology , Mice , Mice, Inbred C57BL , Schistosoma mansoni/physiology , Schistosomiasis mansoni/genetics , Schistosomiasis mansoni/parasitology , Spleen/immunology , T-Lymphocytes, Regulatory/parasitology , Th2 Cells/immunology
3.
PLoS Pathog ; 9(3): e1003215, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23516361

ABSTRACT

The suppression of protective Type 2 immunity is a principal factor driving the chronicity of helminth infections, and has been attributed to a range of Th2 cell-extrinsic immune-regulators. However, the intrinsic fate of parasite-specific Th2 cells within a chronic immune down-regulatory environment, and the resultant impact such fate changes may have on host resistance is unknown. We used IL-4gfp reporter mice to demonstrate that during chronic helminth infection with the filarial nematode Litomosoides sigmodontis, CD4(+) Th2 cells are conditioned towards an intrinsically hypo-responsive phenotype, characterised by a loss of functional ability to proliferate and produce the cytokines IL-4, IL-5 and IL-2. Th2 cell hypo-responsiveness was a key element determining susceptibility to L. sigmodontis infection, and could be reversed in vivo by blockade of PD-1 resulting in long-term recovery of Th2 cell functional quality and enhanced resistance. Contrasting with T cell dysfunction in Type 1 settings, the control of Th2 cell hypo-responsiveness by PD-1 was mediated through PD-L2, and not PD-L1. Thus, intrinsic changes in Th2 cell quality leading to a functionally hypo-responsive phenotype play a key role in determining susceptibility to filarial infection, and the therapeutic manipulation of Th2 cell-intrinsic quality provides a potential avenue for promoting resistance to helminths.


Subject(s)
Cytokines/metabolism , Filariasis/immunology , Filarioidea/immunology , Th2 Cells/immunology , Animals , B7-H1 Antigen/metabolism , Cytokines/analysis , Disease Susceptibility , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Female , Filariasis/parasitology , Flow Cytometry , Humans , Lymphocyte Activation , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Phenotype , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th2 Cells/metabolism
4.
Eur J Immunol ; 43(3): 705-15, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23319295

ABSTRACT

Foxp3(+) regulatory T (Treg) cells are key immune regulators during helminth infections, and identifying the mechanisms governing their induction is of principal importance for the design of treatments for helminth infections, allergies and autoimmunity. Little is yet known regarding the co-stimulatory environment that favours the development of Foxp3(+) Treg-cell responses during helminth infections. As recent evidence implicates the co-stimulatory receptor ICOS in defining Foxp3(+) Treg-cell functions, we investigated the role of ICOS in helminth-induced Foxp3(+) Treg-cell responses. Infection of ICOS(-/-) mice with Heligmosomoides polygyrus or Schistosoma mansoni led to a reduced expansion and maintenance of Foxp3(+) Treg cells. Moreover, during H. polygyrus infection, ICOS deficiency resulted in increased Foxp3(+) Treg-cell apoptosis, a Foxp3(+) Treg-cell specific impairment in IL-10 production, and a failure to mount putatively adaptive Helios(-) Foxp3(+) Treg-cell responses within the intestinal lamina propria. Impaired lamina propria Foxp3(+) Treg-cell responses were associated with increased production of IL-4 and IL-13 by CD4(+) T cells, demonstrating that ICOS dominantly downregulates Type 2 responses at the infection site, sharply contrasting with its Type 2-promoting effects within lymphoid tissue. Thus, ICOS regulates Type 2 immunity in a tissue-specific manner, and plays a key role in driving Foxp3(+) Treg-cell expansion and function during helminth infections.


Subject(s)
Forkhead Transcription Factors/metabolism , Helminthiasis/genetics , Helminthiasis/immunology , Inducible T-Cell Co-Stimulator Protein/genetics , Interleukin-10/biosynthesis , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Apoptosis/genetics , Female , Inducible T-Cell Co-Stimulator Protein/deficiency , Inducible T-Cell Co-Stimulator Protein/metabolism , Male , Mice , Mice, Knockout , Mucous Membrane/immunology , Mucous Membrane/parasitology , Nematospiroides dubius/immunology , Th2 Cells/immunology
5.
Trends Immunol ; 33(4): 181-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22398370

ABSTRACT

Helminth parasites survive through a combination of parasite longevity, repeated re-infection and selective immune suppression to prevent protective Th2 responses. To counteract helminth-induced immunosuppression, and to induce long-term immunological memory, understanding of the multiple regulatory pathways within the T cell compartment is needed. Extrinsic inhibition by regulatory T cells is a key element of Th2 suppression. In addition, Th2 cells in chronic regulatory environments become functionally impaired, indicating cell-intrinsic regulation, which compromises protective Th2 memory. We discuss these pathways and consider the potential for reversing unresponsiveness through stimulatory signals or replacement by new responder populations. Future vaccine or therapeutic strategies should aim to minimize extrinsic regulatory effects and simultaneously negate Th2 anergy to drive effector responses into a long-term functionally competent state.


Subject(s)
Helminthiasis/immunology , T-Lymphocytes/immunology , Adaptation, Biological , Animals , Cell Lineage , Forkhead Transcription Factors/immunology , Helminthiasis/parasitology , Humans , Immunologic Memory , T-Lymphocytes/cytology , T-Lymphocytes/parasitology
6.
J Immunol ; 187(3): 1411-20, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21705620

ABSTRACT

The immune suppression that characterizes human helminth infections can hinder the development of protective immunity or help to reduce pathogenic inflammation. Signaling through the T cell costimulator glucocorticoid-induced TNFR-related protein (GITR) counteracts immune downregulation by augmenting effector T cell responses and abrogating suppression by Foxp3(+) regulatory T cells. Thus, superphysiological Ab-mediated GITR costimulation represents a novel therapy for promoting protective immunity toward parasitic helminths, whereas blocking physiological GITR-GITR ligand (GITRL) interactions may provide a mechanism for dampening pathogenic Th2 inflammation. We investigated the superphysiological and physiological roles of the GITR-GITRL pathway in the development of protective and pathogenic Th2 responses in murine infection models of filariasis (Litomosoides sigmodontis) and schistosomiasis (Schistosoma mansoni). Providing superphysiological GITR costimulation using an agonistic anti-GITR mAb over the first 12 d of L. sigmodontis infection initially increased the quantity of Th2 cells, as well as their ability to produce Th2 cytokines. However, as infection progressed, the Th2 responses reverted to normal infection levels, and parasite killing remained unaffected. Despite the Th2-promoting role of superphysiological GITR costimulation, Ab-mediated blockade of the GITR-GITRL pathway did not affect Th2 cell priming or maintenance during L. sigmodontis infection. Blockade of GITR-GITRL interactions during the acute egg phase of S. mansoni infection resulted in reduced Th2 responses, but this effect was confined to the spleen and did not lead to changes in liver pathology. Thus, although superphysiological GITR costimulation can therapeutically enhance Th2 responses, physiological GITR-GITRL interactions are not required for the development of Th2-mediated resistance or pathology in murine models of filariasis and schistosomiasis.


Subject(s)
Filarioidea/immunology , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Lymphocyte Activation/immunology , Schistosoma mansoni/immunology , Th2 Cells/immunology , Th2 Cells/parasitology , Tumor Necrosis Factors/metabolism , Animals , Female , Filariasis/immunology , Filariasis/pathology , Filariasis/therapy , Glucocorticoid-Induced TNFR-Related Protein/genetics , Glucocorticoid-Induced TNFR-Related Protein/physiology , Humans , Immunity, Innate/genetics , Ligands , Lymphocyte Activation/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/pathology , Schistosomiasis mansoni/therapy , Th2 Cells/pathology , Tumor Necrosis Factors/genetics
7.
Eur J Immunol ; 39(1): 192-206, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19089814

ABSTRACT

Human helminth infections are synonymous with impaired immune responsiveness indicating suppression of host immunity. Using a permissive murine model of filariasis, Litomosoides sigmodontis infection of inbred mice, we demonstrate rapid recruitment and increased in vivo proliferation of CD4(+)Foxp3(+) Treg cells upon exposure to infective L3 larvae. Within 7 days post-infection this resulted in an increased percentage of CD4(+)T cells at the infection site expressing Foxp3. Antibody-mediated depletion of CD25(+) cells prior to infection to remove pre-existing 'natural' CD4(+)CD25(+)Foxp3(+) Treg cells, while not affecting initial larval establishment, significantly reduced the number of adult parasites recovered 60 days post-infection. Anti-CD25 pre-treatment also impaired the fecundity of the surviving female parasites, which had reduced numbers of healthy eggs and microfilaria within their uteri, translating to a reduced level of blood microfilaraemia. Enhanced parasite killing was associated with augmented in vitro production of antigen-specific IL-4, IL-5, IL-13 and IL-10. Thus, upon infection filarial larvae rapidly provoke a CD4(+)Foxp3(+) Treg-cell response, biasing the initial CD4(+) T-cell response towards a regulatory phenotype. These CD4(+)Foxp3(+) Treg cells are predominantly recruited from the 'natural' regulatory pool and act to inhibit protective immunity over the full course of infection.


Subject(s)
Filariasis/immunology , Filarioidea/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD4 Antigens/immunology , Cytokines/biosynthesis , Cytokines/immunology , Female , Filariasis/parasitology , Forkhead Transcription Factors/immunology , Larva/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
8.
Diabetes Metab Res Rev ; 23(3): 169-83, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17103489

ABSTRACT

During the last decades, the incidence of type 1 diabetes (T1D) has increased significantly, reaching percentages of 3% annually worldwide. This increase suggests that besides genetical factors environmental perturbations (including viral infections) are also involved in the pathogenesis of T1D. T1D has been associated with viral infections including enteroviruses, rubella, mumps, rotavirus, parvovirus and cytomegalovirus (CMV). Although correlations between clinical presentation with T1D and the occurrence of a viral infection that precedes the development of overt disease have been recognized, causalities between viruses and the diabetogenic process are still elusive and difficult to prove in humans. The use of experimental animal models is therefore indispensable, and indeed more insight in the mechanism by which viruses can modulate diabetogenesis has been provided by studies in rodent models for T1D such as the biobreeding (BB) rat, nonobese diabetic (NOD) mouse or specific transgenic mouse strains. Data from experimental animals as well as in vitro studies indicate that various viruses are clearly able to modulate the development of T1D via different mechanisms, including direct beta-cell lysis, bystander activation of autoreactive T cells, loss of regulatory T cells and molecular mimicry. Data obtained in rodents and in vitro systems have improved our insight in the possible role of viral infections in the pathogenesis of human T1D. Future studies will hopefully reveal which human viruses are causally involved in the induction of T1D and this knowledge may provide directions on how to deal with viral infections in diabetes-susceptible individuals in order to delay or even prevent the diabetogenic process.


Subject(s)
Diabetes Mellitus, Type 1/etiology , Virus Diseases/complications , Animals , Cardiovirus Infections/complications , Cytomegalovirus Infections/complications , Diabetes Mellitus, Experimental/genetics , Encephalomyocarditis virus , Enterovirus Infections/complications , Genetic Predisposition to Disease , Humans , Mice , Mice, Inbred NOD , Mice, Transgenic , Mumps/complications , Parvoviridae Infections/complications , Rats , Rats, Inbred BB , Rotavirus Infections/complications
9.
Clin Dev Immunol ; 10(2-4): 133-9, 2003.
Article in English | MEDLINE | ID: mdl-14768944

ABSTRACT

BACKGROUND: As one of the natural perturbants, infection with cytomegalovirus (CMV) is believed to play a role in the development of Type I diabetes. Using the DP-BB rat model for autoimmune diabetes, we here report about possible mechanisms responsible for R(at)CMV-induced accelerated onset of diabetes. METHODS: Rats were i.p. infected with 2 x 10(6) plaque forming units (pfu) RCMV and followed for diabetes development. Presence of RCMV antigens and DNA was analyzed by immunohistochemistry and PCR on pancreatic tissue and isolated islets. The effect of viral infection on peritoneal macrophages (pMphi) and diabetes development was studied by analyzing numbers of pMphi, virus permissiveness and by depletion of this subset by peritoneal lavage. RESULTS: RCMV accelerated onset of diabetes without infecting pancreatic islets. Immunohistochemistry and PCR on pancreas and isolated islets indicated that islets are non-permissive for RCMV. Infection results in an influx of pMphi 1 day p.i. of which approximately 0.05% showed signs of reproductive infection. Depletion of pMphi on days 1-3 p.i. completely counteracted the accelerating effect of RCMV. INTERPRETATION: RCMV accelerates onset of diabetes without infecting pancreatic islets. pMphi might function as an carriage to disseminate virus to the pancreas where they enhance activation of autoreactive T cells resulting in accelerated onset of diabetes.


Subject(s)
Cytomegalovirus/physiology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/virology , Macrophages, Peritoneal/immunology , Animals , Blood Cell Count , Diabetes Mellitus, Type 1/pathology , Immunohistochemistry , Macrophages, Peritoneal/cytology , Pancreas/pathology , Pancreas/virology , Rats , Rats, Inbred BB
10.
Clin Dev Immunol ; 10(2-4): 153-60, 2003.
Article in English | MEDLINE | ID: mdl-14768946

ABSTRACT

BACKGROUND: Viral infections are thought to play a role in the development of autoimmune diseases like type 1 diabetes. In this study we investigated the effect of Rat Cytomegalovirus (RCMV) infection on cellular immunity in a well-defined animal model for diabetes, the Biobreeding (BB) rat. METHODS: Diabetes prone (DP)- and Diabetes resistant (DR)-BB rats were infected with 2 x 10(6) plaque forming units (pfu) RCMV. Diabetes development was monitored by frequent blood-glucose analysis. Effects of RCMV on CD4+, CD8+ and Vbeta-TCR+ T-cell subsets were measured in vivo, and in vitro after restimulation with RCMV-infected fibroblasts. Proliferative capacity was determined by 3H-Thymidine incorporation. RESULTS: RCMV-infection resulted in a significant acceleration of diabetes onset in DP-BB rats (p = 0.003). Percentages CD4+ and CD8+ T-cells were not affected in vivo. In vitro, RCMV-restimulation resulted in a decreased CD4+/CD8+ blastoid T-cell ratio compared to ConA (p = 0.00028). Furthermore, RCMV-restimulation resulted in a strong RCMV-specific proliferation, which comprises about 50% of the response triggered by ConA. Vbeta-TCR percentages did not change upon RCMV-infection or RCMV-restimulation. INTERPRETATION: RCMV-restimulation of splenic T-cells in vitro resulted in a strong RCMV-specific proliferation, probably also including autoreactive T-cells. In vivo, this polyclonal response might be involved in the observed accelerated diabetes development in DP-BB rats upon RCMV-infection.


Subject(s)
Cytomegalovirus Infections/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/virology , Disease Models, Animal , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Cytomegalovirus/physiology , Cytomegalovirus Infections/virology , Female , Male , Rats , Spleen/cytology , Spleen/immunology , Spleen/virology , T-Lymphocyte Subsets/immunology
11.
Am J Respir Cell Mol Biol ; 27(2): 244-9, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12151317

ABSTRACT

The aim of this study was to investigate whether the effect of bacillus Calmette-Guérin (BCG) immunization on ovalbumin-induced allergic inflammation in a rat model depends on the genetic predisposition to react with a T helper cell (Th) 2-type cytokine response. This study was performed in an inbred Th2-predisposed "asthma prone" rat strain (brown Norway [BN]) and in an outbred nonpredisposed strain (Sprague Dawley [SD]), to differentiate between genetic and environmental factors. BCG decreased numbers of lung eosinophils and macrophages in the SD rat. This effect was not seen in the BN rat. In the BN rat, but not in the SD rat, BCG downregulated levels of total serum IgE. No significant differences were found with respect to frequencies of IFNgamma- or interleukin-4-producing cells in the lung in both rat strains. These results indicate that the degree and pathway of immunomodulatory effect of BCG in two genetically different rat strains is dependent on the genetic predisposition to develop a Th2-type response. Therefore, differences in genotype in relation to environment may result in difference in involvement of contributing pathogenic factors and thus different responsiveness to therapeutic strategies.


Subject(s)
BCG Vaccine/immunology , Inflammation/immunology , Th2 Cells/immunology , Animals , BCG Vaccine/genetics , Disease Models, Animal , Eosinophils/immunology , Genetic Predisposition to Disease , Immunoglobulin E/blood , Immunoglobulin E/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Lung/cytology , Macrophages/immunology , Macrophages/metabolism , Male , Ovalbumin/immunology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...