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










Publication year range
1.
Eur Heart J Open ; 3(6): oead094, 2023 Nov.
Article in English | MEDLINE | ID: mdl-38025652

ABSTRACT

Aims: Invariant natural killer T (iNKT) cells, a T cell subset that is CD1d-restricted and expresses a semi-invariant T cell receptor, have been proposed to contribute to dyslipidaemia-driven cardiovascular disease due to their ability to specifically recognize lipid antigens. Studies in mice have attributed pro-atherogenic properties to iNKT cells, but studies in humans investigating associations of iNKT cells with incident coronary events (CE) are lacking. Methods and results: Here, we used flow cytometry to enumerate circulating iNKT cells (CD3+ CD1d-PBS57-Tetramer+) in a case-control cohort nested within the prospective population-based Malmö Diet and Cancer Study (n = 416) to explore associations with incident first-time CE during a median follow-up of 14 years. We found a significant inverse association between CD4- and CD8- double negative (DN) iNKT cells and incident CE, with an odds ratio of 0.62 [95% confidence interval (CI) 0.38-0.99; P = 0.046] comparing the highest vs. the lowest tertile of DN iNKT cells. The association remained significant after adjustment for cardiovascular risk factors with an odds ratio of 0.57 (95% CI 0.33-0.99; P = 0.046). In contrast, total iNKT cells were not significantly associated with incident CE after adjustment, with an odds ratio of 0.74 (95% CI 0.43-1.27; P = 0.276). Conclusion: Our findings indicate that animal studies suggesting an atherosclerosis-promoting role for iNKT cells may not translate to human cardiovascular disease as our data show an association between high circulating numbers of DN iNKT cells and decreased risk of incident CE.

2.
Arterioscler Thromb Vasc Biol ; 40(2): 426-436, 2020 02.
Article in English | MEDLINE | ID: mdl-31801374

ABSTRACT

OBJECTIVE: CD4+CD28null T cells have been shown to be associated with recurrent coronary events and suggested as potential biomarker and therapeutic target. It is unknown whether CD4+CD28null T cells associate with first-time cardiovascular events. We examined CD4+CD28null T cells in a prospective population-based cohort and in patients with advanced atherosclerosis. Approach and Results: CD4+CD28null T cells were quantified in 272 individuals experiencing a first-time coronary event during up to 17 years of follow-up and 272 age- and sex-matched controls in a case-control study, nested within the population-based Malmö Diet and Cancer study. The highest tertile of CD4+CD28null T cells was associated with a lower incidence of first-time coronary events compared with the lowest tertile (odds ratio, 0.48 [95% CI, 0.29-0.79], P=0.004) when adjusting for Framingham risk factors. This association remained significant for events recorded after >9 years of follow-up, when most coronary events occurred, but not during the first 9 years of follow-up, despite similar odds ratio. Additionally, we analyzed CD4+CD28null T cells in 201 patients with advanced atherosclerosis undergoing carotid endarterectomy. The adjusted hazard ratio for cardiovascular events in patients with advanced atherosclerosis was 2.11 (95% CI, 1.10-4.05, P=0.024), comparing the highest with the lowest CD4+CD28null T-cell tertile. CONCLUSIONS: Our findings reveal complex associations between CD4+CD28null T cells and cardiovascular disease. Although we confirm the reported positive associations with an adverse prognosis in patients with already established disease, the opposite associations with first-time coronary events in the population-based cohort may limit the clinical use of CD4+CD28null T cells.


Subject(s)
Atherosclerosis/blood , CD28 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Coronary Artery Disease/blood , Coronary Vessels/diagnostic imaging , Forecasting , Atherosclerosis/epidemiology , Atherosclerosis/immunology , Case-Control Studies , Coronary Artery Disease/epidemiology , Coronary Artery Disease/immunology , Female , Flow Cytometry , Follow-Up Studies , Humans , Incidence , Lymphocyte Count , Male , Middle Aged , Population Surveillance , Prospective Studies , Sweden/epidemiology , T-Lymphocytes/immunology
3.
Stem Cells ; 32(5): 1110-23, 2014 May.
Article in English | MEDLINE | ID: mdl-24302539

ABSTRACT

The identity of the cells that contribute to brain tumor structure and progression remains unclear. Mesenchymal stem cells (MSCs) have recently been isolated from normal mouse brain. Here, we report the infiltration of MSC-like cells into the GL261 murine glioma model. These brain tumor-derived mesenchymal stem cells (BT-MSCs) are defined with the phenotype (Lin-Sca-1+CD9+CD44+CD166+/-) and have multipotent differentiation capacity. We show that the infiltration of BT-MSCs correlates to tumor progression; furthermore, BT-MSCs increased the proliferation rate of GL261 cells in vitro. For the first time, we report that the majority of GL261 cells expressed mesenchymal phenotype under both adherent and sphere culture conditions in vitro and that the non-MSC population is nontumorigenic in vivo. Although the GL261 cell line expressed mesenchymal phenotype markers in vitro, most BT-MSCs are recruited cells from host origin in both wild-type GL261 inoculated into green fluorescent protein (GFP)-transgenic mice and GL261-GFP cells inoculated into wild-type mice. We show the expression of chemokine receptors CXCR4 and CXCR6 on different recruited cell populations. In vivo, the GL261 cells change marker profile and acquire a phenotype that is more similar to cells growing in sphere culture conditions. Finally, we identify a BT-MSC population in human glioblastoma that is CD44+CD9+CD166+ both in freshly isolated and culture-expanded cells. Our data indicate that cells with MSC-like phenotype infiltrate into the tumor stroma and play an important role in tumor cell growth in vitro and in vivo. Thus, we suggest that targeting BT-MSCs could be a possible strategy for treating glioblastoma patients.


Subject(s)
Brain Neoplasms/pathology , Brain/pathology , Glioma/pathology , Mesenchymal Stem Cells/pathology , Activated-Leukocyte Cell Adhesion Molecule/metabolism , Animals , Brain/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Differentiation , Cell Line, Tumor , Cells, Cultured , Disease Progression , Flow Cytometry , Glioma/genetics , Glioma/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Hyaluronan Receptors/metabolism , Immunophenotyping , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Mice , Mice, Transgenic , Microscopy, Confocal , Multipotent Stem Cells/metabolism , Multipotent Stem Cells/pathology , Receptors, CXCR/metabolism , Receptors, CXCR4/metabolism , Receptors, CXCR6 , Survival Analysis , Tetraspanin 29/metabolism
4.
J Neurosci ; 33(35): 14231-45, 2013 Aug 28.
Article in English | MEDLINE | ID: mdl-23986257

ABSTRACT

Glioblastoma multiforme (GBM) is the most aggressive form of brain tumor. In general, tumor growth requires disruption of the tissue microenvironment, yet how this affects glioma progression is unknown. We studied program death-ligand (PD-L)1 in neurons and gliomas in tumors from GBM patients and associated the findings with clinical outcome. Remarkably, we found that upregulation of PD-L1 by neurons in tumor-adjacent brain tissue (TABT) associated positively with GBM patient survival, whereas lack of neuronal PD-L1 expression was associated with high PD-L1 in tumors and unfavorable prognosis. To understand the molecular mechanism of PD-L1 signaling in neurons, we investigated PD-L1 function in cerebellar and cortical neurons and its impact on gliomas. We discovered that neuronal PD-L1-induced caspase-dependent apoptosis of glioma cells. Because interferon (IFN)-ß induces PD-L1 expression, we studied the functional consequences of neuronal Ifnb gene deletion on PD-L1 signaling and function. Ifnb-/- neurons lacked PD-L1 and were defective in inducing glioma cell death; this effect was reversed on PD-L1 gene transfection. Ifnb-/- mice with intracerebral isografts survived poorly. Similar to the observations in GBM patients, better survival in wild-type mice was associated with high neuronal PD-L1 in TABT and downregulation of PD-L1 in tumors, which was defective in Ifnb-/- mice. Our data indicated that neuronal PD-L1 signaling in brain cells was important for GBM patient survival. Reciprocal PD-L1 regulation in TABT and tumor tissue could be a prognostic biomarker for GBM. Understanding the complex interactions between tumor and adjacent stromal tissue is important in designing targeted GBM therapies.


Subject(s)
B7-H1 Antigen/genetics , Biomarkers, Tumor/genetics , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Neurons/metabolism , Adult , Aged , Animals , Apoptosis , B7-H1 Antigen/metabolism , Biomarkers, Tumor/metabolism , Brain Neoplasms/diagnosis , Cerebellum/pathology , Cerebral Cortex/pathology , Female , Gene Deletion , Gene Expression Regulation, Neoplastic , Glioblastoma/diagnosis , Humans , Interferon-beta/genetics , Interferon-beta/metabolism , Male , Mice , Middle Aged , Prognosis
5.
Cancer Immunol Immunother ; 61(8): 1191-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22213142

ABSTRACT

Glioblastoma multiforme is the most common and aggressive malignant brain tumor in humans, and the prognosis is very poor despite conventional therapy. Immunotherapy represents a novel treatment approach, but the effect is often weakened by release of immune-suppressive molecules such as prostaglandins. In the current study, we investigated the effect of immunotherapy with irradiated interferon-γ (IFN-γ)-secreting tumor cells and administration of the selective cyclooxygenase-2 (COX-2) inhibitor parecoxib as treatment of established rat brain tumors. COX-2 inhibition and immunotherapy significantly enhanced the long-term cure rate (81% survival) compared with immunotherapy alone (19% survival), and there was a significant increase in plasma IFN-γ levels in animals treated with the combined therapy, suggesting a systemic T helper 1 immune response. COX-2 inhibition alone, however, did neither induce cure nor prolonged survival. The tumor cells were identified as the major source of COX-2 both in vivo and in vitro, and unmodified tumor cells produced prostaglandin E(2) in vitro, while the IFN-γ expressing tumor cells secreted significantly lower levels. In conclusion, we show that immunotherapy of experimental brain tumors is greatly potentiated when combined with COX-2 inhibition. Based on our results, the clinically available drug parecoxib may be added to immunotherapy against human brain tumors. Furthermore, the discovery that IFN-γ plasma levels can be used to determine the ongoing in vivo immune response has translational potential.


Subject(s)
Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/metabolism , Glioblastoma/therapy , Immunotherapy, Adoptive/methods , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/therapy , Animals , Brain Neoplasms/enzymology , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Combined Modality Therapy , Disease Models, Animal , Flow Cytometry , Glioblastoma/enzymology , Glioblastoma/immunology , Immunohistochemistry , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Isoxazoles/pharmacology , Male , Neoplasms, Experimental/immunology , Rats , Rats, Inbred F344
6.
Curr Pharm Des ; 16(4): 428-30, 2010.
Article in English | MEDLINE | ID: mdl-20236071

ABSTRACT

Malignant gliomas bear the most dismal prognosis of all human cancers despite the progress in therapy of many other tumors. The search for alternative and complementary treatments has therefore a high priority. Emerging knowledge of the dual and diverging role of nitric oxide in glioma biology has focused on possibilities to achieve anti-glioma effects by modulation of nitric oxide (NO) release and function in these tumors. NO has been shown to influence proliferation of glioma cells, vascular function in gliomas, invasive capacity of gliomas, effects of chemo and radiotherapy and also immune reactivity against these tumors. The mechanisms behind the reported diverse and dual effects of NO in glioma biology are multiple. Some of the diversity can be explained by different experimental setups as in vitro versus in vivo models but the cellular sources, timing, absolute levels and gradients play a decisive role for the effects of NO on glioma biology. Current research in this field is hampered by the lack of inhibitors and donors approved for clinical use.


Subject(s)
Glioma/metabolism , Nitric Oxide/metabolism , Animals , Cell Proliferation/drug effects , Glioma/pathology , Glioma/therapy , Humans , Nitric Oxide/pharmacology , Nitric Oxide/therapeutic use
7.
J Immunother ; 32(6): 593-601, 2009.
Article in English | MEDLINE | ID: mdl-19483650

ABSTRACT

Immunotherapy of malignant primary brain tumors holds the potential to improve the dismal prognosis after current clinical therapy. Although immunotherapy of experimental gliomas has been demonstrated to have the capacity to cure intracerebral tumors no convincing effects of immunotherapy have been shown in clinical trials. One reason for this could be that some of the models used do not display full features of human glioblastomas. The N29 rat gliomas exhibited all the histologic features of human glioblastoma multiforme including nuclear atypia, mitotic figures, necrosis, and diffuse infiltration into the normal brain tissue. Surprisingly, immunotherapy with autologous interferon gamma producing tumor cells against preestablished intracerebral N29 tumors yielded a higher cure rate than immunotherapy against less invasive tumors. Furthermore, when immunizations were postponed until day 5 after tumor establishment 50% of the animals survived. When immunizations were postponed until day 11 after tumor establishment no glioma-bearing animals were cured but survival was significantly prolonged. The superior effect of immunotherapy in the invasive N29 model compared with the less invasive tumors could depend on combined effects of up-regulation of major histocompatibility complex I and induction of major histocompatibility complex II plus CD80 after transfection and irradiation of the tumor cells used for immunizations. This study demonstrates that immunotherapy against experimental brain tumors indeed is feasible even against highly invasive and established tumors. These results strengthen the translational potential of immunotherapy against malignant brain tumors.


Subject(s)
Central Nervous System Neoplasms/therapy , Glioma/therapy , Immunotherapy, Adoptive/methods , Interferon-gamma/immunology , Animals , Apoptosis/immunology , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , Cell Line, Tumor , Central Nervous System Neoplasms/immunology , Central Nervous System Neoplasms/pathology , Glioma/immunology , Glioma/pathology , Major Histocompatibility Complex/immunology , Male , Rats , Rats, Inbred F344
8.
Int J Cancer ; 124(3): 630-7, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-18972433

ABSTRACT

We were the first to demonstrate that combined immunotherapy with GM-CSF producing GL261 cells and recombinant IFNgamma of preestablished GL261 gliomas could cure 90% of immunized mice. To extend these findings and to uncover the underlying mechanisms, the ensuing experiments were undertaken. We hypothesized that immunizations combining both GM-CSF and IFNgamma systemically would increase the number of immature myeloid cells, which then would mature and differentiate into dendritic cells (DCs) and macrophages, thereby augmenting tumor antigen presentation and T-cell activation. Indeed, the combined therapy induced a systemic increase of both immature and mature myeloid cells but also an increase in T regulatory cells (T-regs). Cytotoxic anti-tumor responses, mirrored by an increase in Granzyme B-positive cells as well as IFNgamma-producing T-cells, were augmented after immunizations with GM-CSF and IFNgamma. We also show that the combined therapy induced a long-term memory with rejection of intracerebral (i.c.) rechallenges. Depletion of T-cells showed that both CD4+ and CD8+ T-cells were essential for the combined GM-CSF and IFNgamma effect. Finally, when immunizations were delayed until day 5 after tumor inoculation, only mice receiving immunotherapy with both GM-CSF and IFNgamma survived. We conclude that the addition of recombinant IFNgamma to immunizations with GM-CSF producing tumor cells increased the number of activated tumoricidal T-cells, which could eradicate established intracerebral tumors. These results clearly demonstrate that the combination of cytokines in immunotherapy of brain tumors have synergistic effects that have implications for clinical immunotherapy of human malignant brain tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/immunology , Glioma/drug therapy , Glioma/immunology , Immunotherapy/methods , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Immunologic Memory/drug effects , Interferon-gamma/administration & dosage , Male , Mice , Mice, Inbred C57BL
9.
J Immunol ; 179(6): 4231-8, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17785863

ABSTRACT

High-grade gliomas are one of the most aggressive human tumors with <1% of patients surviving 5 years after surgery. Immunotherapy could offer a possibility to eradicate remnant tumor cells after conventional therapy. Experimental immunotherapy can induce partial cure of established intracerebral tumors in several rodent models. One reason for the limited therapeutic effects could be immunosuppression induced by both the growing tumor and the induced immune reaction. NO has been implicated in tumor-derived immune suppression in tumor-bearing hosts, and unspecific inhibitors of NO synthase have been shown to boost antitumor immunity. In this study, we show that the inducible NO synthase (iNOS)-specific inhibitor mercaptoethylguanidine (MEG) superiorly enhanced lymphocyte reactivity after polyclonal stimulation compared with the iNOS-specific inhibitor L-NIL and the unspecific NO synthase inhibitor L-NAME. Both iNOS inhibitors increased the number and proliferation of T cells but not of B cells. When combined during postimmunization with IFN-gamma-secreting N32 rat glioma cells of rats harboring intracerebral tumors, only MEG increased the cure rate. However, this was only achieved when MEG was administered after immunizations. These findings implicate that NO has both enhancing and suppressive effects after active immunotherapy.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Glioma/drug therapy , Glioma/metabolism , Guanidines/administration & dosage , Immunotherapy, Adoptive , Interferon-gamma/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Animals , Brain Neoplasms/immunology , Brain Neoplasms/mortality , Cell Line, Tumor , Cell Proliferation/drug effects , Combined Modality Therapy , Enzyme Inhibitors/administration & dosage , Glioma/immunology , Glioma/mortality , Immunization , Interferon-gamma/biosynthesis , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/pathology , Lysine/administration & dosage , Lysine/analogs & derivatives , Nitric Oxide Synthase Type II/physiology , Rats , Rats, Inbred F344 , Spleen/cytology , Spleen/drug effects , Spleen/immunology , Survival , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology
10.
Int J Cancer ; 120(1): 75-80, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17044023

ABSTRACT

Glioblastoma multiforme is the most common malignant primary brain tumor and also one of the most therapy-resistant tumors. Because of the dismal prognosis, various therapies modulating the immune system have been developed in experimental models. Previously, we have shown a 37-70% cure in a rat glioma model where rats were peripherally immunized with tumor cells producing IFNgamma. On the basis of these results, we wanted to investigate whether a combination of GM-CSF and IFNgamma could improve the therapeutic effect in a mouse glioma model, GL261 (GL-wt). Three biweekly intraperitoneal (i.p.) immunizations with irradiated GM-CSF-transduced GL261 cells (GL-GM) induced a 44% survival in mice with intracranial glioma. While treatment of GL-wt and GL-GM with IFNgamma in vitro induced upregulation of MHC I and MHC II on the tumor cells, it could not enhance survival after immunization. However, immunizations with GL-GM combined with recombinant IFNgamma at the immunization site synergistically enhanced survival with a cure rate of 88%. Tumors from mice receiving only 1 immunization on Day 10 after tumor inoculation were sectioned on Day 20 for analysis of leukocyte infiltration. Tumor volume was reduced and the infiltration of macrophages was denser in mice immunized with GL-GM combined with IFNgamma compared with that of both wildtype and nonimmunized mice. To our knowledge, this is the first study to demonstrate a synergy between GM-CSF and IFNgamma in experimental immunotherapy of tumors, by substantially increasing survival as well as inducing a potent anti-tumor response after only 1 postponed immunization.


Subject(s)
Antiviral Agents/therapeutic use , Brain Neoplasms/drug therapy , Glioma/drug therapy , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Immunotherapy , Interferon-gamma/therapeutic use , Animals , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Drug Synergism , Drug Therapy, Combination , Genes, MHC Class I/physiology , Genes, MHC Class II/physiology , Genetic Therapy , Genetic Vectors , Glioma/immunology , Glioma/pathology , Immunization , Immunoenzyme Techniques , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Recombinant Proteins , Survival Rate , Transduction, Genetic , Tumor Cells, Cultured/radiation effects
11.
Clin Cancer Res ; 12(15): 4714-9, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16899622

ABSTRACT

PURPOSE: Although there is a need to enhance the therapeutic efficiency in cancer by combining immunotherapeutic procedures with other therapy, combination with chemotherapy is complicated due to immunosuppressive effects of most chemotherapeutic drugs. The purpose of this investigation was to study whether combining tumor cell immunization with the vascular targeting drug combretastatin A4 phosphate (CA4P) would enhance tumor retardation and/or affect the antitumor immune response. EXPERIMENTAL DESIGN: Rats with intrahepatic colon carcinoma were immunized weekly with IL-18/IFNgamma-transfected tumor cells, starting day 9, and were treated with a low-dose CA4P (2 mg/kg, 5 days a week starting day 7). The effect of CA4P was studied on tumor growth and on immune reactivity in vitro. RESULTS: Rats with preexisting tumor, immunized and treated with low-dose CA4P, had a significantly retarded tumor growth compared with rats receiving CA4P or immunization alone. Splenocytes from rats treated with this combination had a significantly enhanced antitumor immune response compared with splenocytes from control rats. Exposure of nonadherent splenocytes to CA4P in vitro did not enhance their proliferation. However, 3-hour pretreatment of adherent splenocytes with 0.3 microg/mL CA4P significantly enhanced proliferation and IFNgamma production of admixed nonadherent splenocytes, partly due to nitric oxide reduction. Combining the nitric oxide synthase inhibitor N-nitro-l-arginine methyl ester with CA4P and immunization further retarded tumor growth. CONCLUSION: Concomitant treatment of rats with progressively growing tumor with immunization and low-dose CA4P significantly enhances the therapeutic effect as compared with either treatment alone and results in an enhanced antitumor immune reactivity.


Subject(s)
Carcinoma/drug therapy , Carcinoma/immunology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/immunology , Stilbenes/administration & dosage , Administration, Oral , Animals , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Dose-Response Relationship, Immunologic , Humans , Injections, Intraperitoneal , Interferon-gamma/biosynthesis , Interferon-gamma/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/immunology , Rats , Rats, Inbred BN , Rats, Inbred WF , Stilbenes/pharmacology , Structure-Activity Relationship , Transplantation, Heterologous , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...