Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 13: 971001, 2022.
Article in English | MEDLINE | ID: mdl-36330506

ABSTRACT

Virtual memory CD8+ T cells (TVM) have been described as cells with a memory-like phenotype but without previous antigen (Ag) exposure. TVM cells have the ability to respond better to innate stimuli rather than by TCR engagement, producing large amounts of interferon gamma (IFNγ) after stimulation with interleukin (IL)-12 plus IL-18. As a result of the phenotypic similarity, TVM cells have been erroneously included in the central memory T cell subset for many years. However, they can now be discriminated via the CD49d receptor, which is up-regulated only on conventional memory T cells (TMEM) and effector T cells (TEFF) after specific cognate Ag recognition by a TCR. In this work we show that systemic expression of IL-12 plus IL-18 induced an alteration in the normal TVM vs TMEM/TEFF distribution in secondary lymphoid organs and a preferential enrichment of TVM cells in the melanoma (B16) and the pancreatic ductal adenocarcinoma (KPC) tumor models. Using our KPC bearing OT-I mouse model, we observed a significant increase in CD8+ T cell infiltrating the tumor islets after IL-12+IL-18 stimulation with a lower average speed when compared to those from control mice. This finding indicates a stronger interaction of T cells with tumor cells after cytokine stimulation. These results correlate with a significant reduction in tumor size in both tumor models in IL-12+IL-18-treated OT-I mice compared to control OT-I mice. Interestingly, the absence of IFNγ completely abolished the high antitumor capacity induced by IL-12+IL-18 expression, indicating an important role for these cytokines in early tumor growth control. Thus, our studies provide significant new information that indicates an important role of TVM cells in the immune response against cancer.


Subject(s)
Interferon-gamma , Neoplasms , Mice , Animals , Interferon-gamma/metabolism , CD8-Positive T-Lymphocytes , Interleukin-18 , Immunologic Memory , Interleukin-12/pharmacology , Cytokines/metabolism , Receptors, Antigen, T-Cell
2.
Immunotherapy ; 14(2): 115-133, 2022 02.
Article in English | MEDLINE | ID: mdl-34783257

ABSTRACT

Aim: The aim of this work is to utilize a gene expression procedure to safely express systemic IL-12 and evaluate its effects in mouse tumor models. Materials & methods: Secondary lymphoid organs and tumors from EL4 and B16 tumor-bearing mice were analyzed by supervised and unsupervised methods. Results: IL-12 cDNA induced systemic IL-12 protein levels lower than the tolerated dose in patients. Control of tumor growth was observed in subcutaneous B16 and EL4 tumors. Systemic IL-12 expression induced a higher frequency of both total tumor-infiltrated CD45+ cells and proliferative IFN-γ+CD8+ T cells along with a lower frequency of CD4+FOXP3+ and CD11b+Gr-1+ cells. Conclusion: This approach characterizes the systemic effects of IL-12, helping to improve treatment of metastases or solid tumors.


Lay abstract IL-12 has emerged as a potent cytokine in mediating antitumor activity in preclinical models of cancer. However, this antitumor response has not yet been translated into the clinic because of toxic side effects. The aim of our work is to analyze the effects of IL-12 in mouse tumor models. We demonstrate that one injection of IL-12 cDNA can induce systemic IL-12 levels in serum even lower than the tolerated dose in patients. At this dose, an efficient control of tumor growth can be observed. We found a higher frequency of both total tumor-infiltrated leukocytes and IFN-γ-producing CD8+ T cells along with a lower frequency of regulatory CD4+FOXP3+ and CD11b+Gr1+ cells. Our work demonstrates that IL-12 cDNA can safely be used to treat cancer.


Subject(s)
Adjuvants, Immunologic/therapeutic use , DNA, Complementary/blood , Interleukin-12/therapeutic use , Lymphoma/drug therapy , Melanoma, Experimental/drug therapy , Animals , Disease Models, Animal , Gene Expression , Interleukin-12/blood , Lymphoma/blood , Lymphoma/immunology , Melanoma, Experimental/blood , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Signal Transduction
3.
PLoS Pathog ; 15(1): e1007456, 2019 01.
Article in English | MEDLINE | ID: mdl-30608984

ABSTRACT

Innate CD8+ T cells express a memory-like phenotype and demonstrate a strong cytotoxic capacity that is critical during the early phase of the host response to certain bacterial and viral infections. These cells arise in the thymus and depend on IL-4 and IL-15 for their development. Even though innate CD8+ T cells exist in the thymus of WT mice in low numbers, they are highly enriched in KO mice that lack certain kinases, leading to an increase in IL-4 production by thymic NKT cells. Our work describes that in C57BL/6 WT mice undergoing a Th1 biased infectious disease, the thymus experiences an enrichment of single positive CD8 (SP8) thymocytes that share all the established phenotypical and functional characteristics of innate CD8+ T cells. Moreover, through in vivo experiments, we demonstrate a significant increase in survival and a lower parasitemia in mice adoptively transferred with SP8 thymocytes from OT I-T. cruzi-infected mice, demonstrating that innate CD8+ thymocytes are able to protect against a lethal T. cruzi infection in an Ag-independent manner. Interestingly, we obtained similar results when using thymocytes from systemic IL-12 + IL-18-treated mice. This data indicates that cytokines triggered during the acute stage of a Th1 infectious process induce thymic production of IL-4 along with IL-15 expression resulting in an adequate niche for development of innate CD8+ T cells as early as the double positive (DP) stage. Our data demonstrate that the thymus can sense systemic inflammatory situations and alter its conventional CD8 developmental pathway when a rapid innate immune response is required to control different types of pathogens.


Subject(s)
Interleukin-15/metabolism , Interleukin-4/metabolism , Thymus Gland/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Cytokines/metabolism , Female , Immunity, Innate/genetics , Interleukin-12/metabolism , Interleukin-15/genetics , Interleukin-18/metabolism , Interleukin-4/genetics , Male , Mice , Mice, Inbred C57BL , Natural Killer T-Cells/immunology , Signal Transduction , Th1 Cells/immunology , Thymocytes/metabolism , Thymus Gland/metabolism , Thymus Gland/pathology
4.
J Neuroimmunol ; 313: 145-151, 2017 12 15.
Article in English | MEDLINE | ID: mdl-28992974

ABSTRACT

Benzodiazepines are psychoactive drugs and some of them also affect immune cells. We here characterized the inflammatory and infiltrating immune cells in the central nervous system (CNS) during the acute phase of experimental autoimmune encephalomyelitis (EAE) in animals treated with Diazepam. Also, we evaluated the expression of Translocator Protein (18kDa) (TSPO), which is a biomarker of neuroinflammatory diseases. The results indicate that Diazepam exerts protective effects on EAE development, decreasing the incidence of the disease and reducing the number of inflammatory cells in CNS, with a concomitant decrease of TSPO levels in brain tissue and CNS inflammatory CD11b+ cells.


Subject(s)
CD11b Antigen/metabolism , Carrier Proteins/metabolism , Central Nervous System/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Hypnotics and Sedatives/therapeutic use , Receptors, GABA-A/metabolism , Animals , Carrier Proteins/genetics , Cytokines/metabolism , Diazepam/therapeutic use , Disease Models, Animal , Lymphocytes/pathology , Macrophages/pathology , Monocytes/pathology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, GABA-A/genetics , Time Factors
5.
PLoS One ; 9(2): e90116, 2014.
Article in English | MEDLINE | ID: mdl-24587231

ABSTRACT

For more than a decade, the cytokine interleukin-12 (IL-12) has been utilized, either alone or in combination with other drugs, as a treatment for cancer. The numerous anti-tumor properties of IL-12 still generate interest in the clinical use of this cytokine, even though it has demonstrated toxicity when administrated systemically. As an approach to overcome this toxicity, numerous laboratories have attempted to induce IL-12 expression at the site of the tumor. However for tumors that are difficult to remove surgically or for the treatment of disseminated metastases, systemic expression of this cytokine still remains as the most efficient method of administration. Nevertheless, finding alternative approaches for the use of IL-12 in the treatment of cancer and unraveling the basis of IL-12-side effects remain a challenge. In the present work we demonstrate that systemic expression of IL-12 through hydrodynamic injection of IL-12 cDNA is able to induce different types of liver lesions associated with a toxic pathology. However we report here that hepatic toxicity is diminished and survival of mice enhanced in the absence of tumor necrosis factor alpha (TNFα). This observation is in contrast to several murine models and clinical trials that postulate interferon gamma (IFNγ) as the main cytokine responsible for IL-12 toxicity. Moreover, our work demonstrates that when IL-12 cDNA is co-injected with IL-18 cDNA or when mice are pre-treated with a low dose of IL-12 cDNA prior to receiving a high dose of IL-12 cDNA, systemic levels of TNFα are almost completely abrogated, resulting in improved survival and less hepatic damage. Importantly, abrogation of TNFα signaling does not affect the strong anti-tumor activity of IL-12. Thus, neutralizing TNFα with antagonists already approved for human use offers a promising approach to abrogate IL-12 side effects during the use of this cytokine for the treatment of cancer.


Subject(s)
DNA, Complementary/administration & dosage , Immunotherapy/methods , Interleukin-12/immunology , Interleukin-18/immunology , Melanoma, Experimental/therapy , Splenic Neoplasms/therapy , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , DNA, Complementary/immunology , Gene Expression , Hydrodynamics , Injections, Intravenous , Interleukin-12/biosynthesis , Interleukin-12/genetics , Interleukin-18/biosynthesis , Interleukin-18/genetics , Liver/drug effects , Liver/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Splenic Neoplasms/immunology , Splenic Neoplasms/pathology , Tail , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
6.
Eur J Immunol ; 42(10): 2644-54, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22740067

ABSTRACT

Mature lymphocyte immigration into the thymus has been documented in mouse, rat, and pig models, and highly increases when cells acquire an activated phenotype. Entrance of peripheral B and T cells into the thymus has been described in healthy and pathological situations. However, it has not been proposed that leukocyte recirculation to the thymus could be a common feature occurring during the early phase of a Th1 inflammatory/infectious process when a large number of peripheral cells acquire an activated phenotype and the cellularity of the thymus is seriously compromised. The data we present here demonstrate that in well-established Th1 models triggered by different types of immunogens, for example, LPS treatment (a bacterial product), Candida albicans infection (a fungus), and after Trypanosoma cruzi infection (a parasite), a large number of mature peripheral B and T cells enter the thymus. This effect is dependent on, but not exclusive of, the available space in the thymus. Our data also demonstrate that MCP-1/CCR2 (where MCP-1 is monocyte chemoattractant protein-1) interaction is responsible for the infiltration of peripheral cells to the thymus in these Th1-inflammatory/infectious situations. Finally, systemic expression of IL-12 and IL-18 produced during the inflammatory process is ultimately responsible for these migratory events.


Subject(s)
B-Lymphocytes/immunology , Candida albicans/immunology , Candidiasis/immunology , Chagas Disease/immunology , Chemokine CCL2/metabolism , Receptors, CCR2/metabolism , Th1 Cells/immunology , Trypanosoma cruzi/immunology , Animals , B-Lymphocytes/microbiology , B-Lymphocytes/parasitology , Cell Movement , Cells, Cultured , Female , Interleukin-12/immunology , Interleukin-18/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Binding , Th1 Cells/microbiology , Th1 Cells/parasitology , Thymus Gland/immunology , Thymus Gland/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...