Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
NPJ Syst Biol Appl ; 10(1): 66, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38858414

ABSTRACT

Cell-cell crosstalk involves simultaneous interactions of multiple receptors and ligands, followed by downstream signaling cascades working through receptors converging at dominant transcription factors, which then integrate and propagate multiple signals into a cellular response. Single-cell RNAseq of multiple cell subsets isolated from a defined microenvironment provides us with a unique opportunity to learn about such interactions reflected in their gene expression levels. We developed the interFLOW framework to map the potential ligand-receptor interactions between different cell subsets based on a maximum flow computation in a network of protein-protein interactions (PPIs). The maximum flow approach further allows characterization of the intracellular downstream signal transduction from differentially expressed receptors towards dominant transcription factors, therefore, enabling the association between a set of receptors and their downstream activated pathways. Importantly, we were able to identify key transcription factors toward which the convergence of multiple receptor signaling pathways occurs. These identified factors have a unique role in the integration and propagation of signaling following specific cell-cell interactions.


Subject(s)
Signal Transduction , Signal Transduction/physiology , Humans , Transcription Factors/metabolism , Transcription Factors/genetics , Single-Cell Analysis/methods , Cell Communication/physiology , Computational Biology/methods , Ligands , Protein Interaction Maps/genetics , Models, Biological
2.
J Leukoc Biol ; 116(2): 424-435, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38478700

ABSTRACT

Eosinophils have been mainly studied in allergic diseases and parasitic infections. Nonetheless, eosinophils accumulate in a variety of solid tumors, including colorectal cancer, where their presence is associated with improved prognosis. Eosinophils can promote antitumor immunity through various mechanisms, including direct cytotoxicity toward tumor cells and promoting T-cell activation. However, the mechanisms by which tumor cells regulate eosinophil activities are largely unknown. Herein, we characterized the potential interactions between eosinophils and colorectal cancer cells using an unbiased transcriptomic and proteomic analyses approach. Human eosinophils were stimulated with colorectal cancer cell conditioned media, containing tumor cell secreted factors from multiple cancer cell lines. RNA sequencing analysis identified a "core" signature consisting of 101 genes that characterize a baseline transcriptional program for the response of human eosinophils to colorectal cancer cells. Among these, the increased expression of IL-3Rα and its ßc chain was identified and validated at the protein level. Secreted factors from tumor cells potentiated IL-3-induced expression of the adhesion molecule CD11a in eosinophils. Combining proteomics analysis of tumor cell secreted factors with RNA sequencing revealed potential ligand-receptor pairs between tumor cells and eosinophils and the potential involvement of the adhesion molecule CD18 and F2RL3/PAR4. Subsequent functional analyses demonstrated that F2RL3/PAR4 suppresses eosinophil migration in response tumor cell secreted factors. These findings add to the growing body of evidence that eosinophils are conditioned by their local microenvironment. Identifying mechanisms by which eosinophils interact with tumor cells could lead to the development of new immunotherapies for colorectal cancer and other solid tumors.


Subject(s)
Colorectal Neoplasms , Eosinophils , Transcriptome , Humans , Colorectal Neoplasms/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Eosinophils/immunology , Eosinophils/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Gene Expression Profiling
3.
Cancer Cell ; 41(2): 235-251.e9, 2023 02 13.
Article in English | MEDLINE | ID: mdl-36638785

ABSTRACT

Cancer immunotherapy critically depends on fitness of cytotoxic and helper T cell responses. Dysfunctional cytotoxic T cell states in the tumor microenvironment (TME) are a major cause of resistance to immunotherapy. Intratumoral myeloid cells, particularly blood-borne myeloids (bbm), are key drivers of T cell dysfunction in the TME. We show here that major histocompatibility complex class II (MHCII)-restricted antigen presentation on bbm is essential to control the growth of brain tumors. Loss of MHCII on bbm drives dysfunctional intratumoral tumor-reactive CD8+ T cell states through increased chromatin accessibility and expression of Tox, a critical regulator of T cell exhaustion. Mechanistically, MHCII-dependent activation of CD4+ T cells restricts myeloid-derived osteopontin that triggers a chronic activation of NFAT2 in tumor-reactive CD8+ T cells. In summary, we provide evidence that MHCII-restricted antigen presentation on bbm is a key mechanism to directly maintain functional cytotoxic T cell states in brain tumors.


Subject(s)
Brain Neoplasms , T-Lymphocytes, Cytotoxic , Humans , Antigen Presentation , CD8-Positive T-Lymphocytes , Histocompatibility Antigens Class II/metabolism , Tumor Microenvironment
4.
Immun Inflamm Dis ; 10(11): e728, 2022 11.
Article in English | MEDLINE | ID: mdl-36301029

ABSTRACT

BACKGROUND: CCAAT/enhancer-binding protein ß (C/EBPß) is a transcription factor known to be involved in macrophage differentiation and function, steatohepatitis and liver fibrosis. METHODS: Immune restricted C/EBPß deficient and control mice were investigated in steady-state and in the CDA-HFD steatohepatitis model. Mice were assessed for weight change, liver biochemical profile, histology and hepatic phagocytes composition. RESULTS: Flow cytometry analysis of hepatic nonparenchymal cells revealed reduced numbers of hepatic monocytes and Kupffer cells and an increase in hepatic MHC class II positive myeloid cells in immune cells restricted C/EBPß deficient mice. Immune-restricted C/EBPß deficiency resulted in decreased weight gain and appearance of mild spontaneous liver inflammation. Nevertheless, In the CDA-HFD steatohepatitis model, immune restricted C/EBPß deficient and proficient mice exhibit similar grade of hepatic steatosis, liver enzymes levels and fibrosis stage. CONCLUSIONS: Immune-restricted C/EBPß deficiency leads to significant alteration in hepatic mononuclear phagocytes composition associated with spontaneous mild hepatitis. Steatohepatitis associated fibrosis is not dependent on C/EBPß expression by immune cells.


Subject(s)
Fatty Liver , Hepatitis , Mice , Animals , Fatty Liver/complications , Liver Cirrhosis/complications , Hepatitis/complications , Gene Expression Regulation
5.
Front Immunol ; 9: 1852, 2018.
Article in English | MEDLINE | ID: mdl-30158929

ABSTRACT

The therapy of primary biliary cholangitis (PBC) has lagged behind other autoimmune diseases despite significant improvements in our understanding of both immunological and molecular events that lead to loss of tolerance to the E2 component of pyruvate dehydrogenase, the immunodominant autoepitope of PBC. It is well known that Ly6Chi monocytes are innate immune cells infiltrating inflammatory sites that are dependent on the expression of C-C motif chemokine receptor 2 (CCR2) for emigration from bone marrow. Importantly, humans with PBC have a circulating monocyte pro-inflammatory phenotype with macrophage accumulation in portal tracts. We have taken advantage of an inducible chemical xenobiotic model of PBC and recapitulated the massive infiltration of monocytes to portal areas. To determine the clinical significance, we immunized both CCR2-deficient mice and controls with 2OA-BSA and noted that CCR2 deficiency is protective for the development of autoimmune cholangitis. Importantly, because of the therapeutic potential, we focused on inhibiting monocyte infiltration through the use of cenicriviroc (CVC), a dual chemokine receptor CCR2/CCR5 antagonist shown to be safe in human trials. Importantly, treatment with CVC resulted in amelioration of all aspects of disease severity including serum total bile acids, histological severity score, and fibrosis stage. In conclusion, our results indicate a major role for Ly6Chi monocytes and for CCR2 in PBC pathogenesis and suggest that inhibition of this axis by CVC should be explored in humans through the use of clinical trials.


Subject(s)
Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Cholangitis/immunology , Cholangitis/metabolism , Monocytes/immunology , Monocytes/metabolism , Receptors, CCR2/metabolism , Animals , Autoimmune Diseases/complications , Autoimmune Diseases/pathology , Biomarkers , Chemokines/metabolism , Cholangitis/complications , Cholangitis/pathology , Disease Models, Animal , Disease Susceptibility , Female , Humans , Imidazoles/pharmacology , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Monocytes/drug effects , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR2/genetics , Sulfoxides , THP-1 Cells
6.
Clin Rev Allergy Immunol ; 52(3): 460-472, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27677500

ABSTRACT

As physiological pathways of intercellular communication produced by all cells, cytokines are involved in the pathogenesis of inflammatory insulitis as well as pivotal mediators of immune homeostasis. Proinflammatory cytokines including interleukins, interferons, transforming growth factor-ß, tumor necrosis factor-α, and nitric oxide promote destructive insulitis in type 1 diabetes through amplification of the autoimmune reaction, direct toxicity to ß-cells, and sensitization of islets to apoptosis. The concept that neutralization of cytokines may be of therapeutic benefit has been tested in few clinical studies, which fell short of inducing sustained remission or achieving disease arrest. Therapeutic failure is explained by the redundant activities of individual cytokines and their combinations, which are rather dispensable in the process of destructive insulitis because other cytolytic pathways efficiently compensate their deficiency. Proinflammatory cytokines are less redundant in regulation of the inflammatory reaction, displaying protective effects through restriction of effector cell activity, reinforcement of suppressor cell function, and participation in islet recovery from injury. Our analysis suggests that the role of cytokines in immune homeostasis overrides their contribution to ß-cell death and may be used as potent immunomodulatory agents for therapeutic purposes rather than neutralized.


Subject(s)
Antibodies, Neutralizing/therapeutic use , Autoimmunity , Diabetes Mellitus, Type 1/therapy , Immunotherapy/methods , Insulin-Secreting Cells/pathology , Apoptosis , Clinical Trials as Topic , Cytokines/immunology , Cytokines/metabolism , Diabetes Mellitus, Type 1/immunology , Homeostasis , Humans , Inflammation Mediators/metabolism , Treatment Failure
7.
Immunology ; 142(3): 465-73, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24601987

ABSTRACT

The non-obese diabetic (NOD) mouse is a prevalent disease model of type 1 diabetes. Immune aberrations that cause and propagate autoimmune insulitis in these mice are being continually debated, with evidence supporting both dominance of effector cells and insufficiency of suppressor mechanisms. In this study we assessed the behaviour of NOD lymphocytes under extreme expansion conditions using adoptive transfer into immunocompromised NOD.SCID (severe combined immunodeficiency) mice. CD4(+)  CD25(+) T cells do not cause islet inflammation, whereas splenocytes and CD4(+)  CD25(-) T cells induce pancreatic inflammation and hyperglycaemia in 80-100% of the NOD.SCID recipients. Adoptively transferred effector T cells migrate to the lymphoid organs and pancreas, proliferate, are activated in the target organ in situ and initiate inflammatory insulitis. Reconstitution of all components of the CD4(+) subset emphasizes the plastic capacity of different cell types to adopt effector and suppressor phenotypes. Furthermore, similar immune profiles of diabetic and euglycaemic NOD.SCID recipients demonstrate dissociation between fractional expression of CD25 and FoxP3 and the severity of insulitis. There were no evident and consistent differences in diabetogenic activity and immune reconstituting activity of T cells from pre-diabetic (11 weeks) and new onset diabetic NOD females. Similarities in immune phenotypes and variable distribution of effector and suppressor subsets in various stages of inflammation commend caution in interpretation of quantitative and qualitative aberrations as markers of disease severity in adoptive transfer experiments.


Subject(s)
Adoptive Transfer , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Immunocompromised Host , Age Factors , Animals , Immunophenotyping , Mice , Mice, Inbred NOD , Mice, SCID , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
8.
Autoimmunity ; 47(2): 105-12, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24328490

ABSTRACT

Immune profiling of non-obese diabetic (NOD) is a widely employed tool to assess the mechanisms of inflammatory insulitis. Our analysis of the female NOD colony revealed similar distribution of lymphoid lineages to wild type mice, and at various ages of prediabetic and diabetic mice. The profiles of mesenteric and pancreatic lymph nodes differ and often change reciprocally due to directed migration of T cells towards the site of inflammation. Significant events in our colony include early decline in CD4(+)CD25(+)CD62L(+) Treg, accompanied by gradual increase in CD4(+)CD25(+)FoxP3(+) Treg in peripheral lymphoid organs and pancreatic infiltrates. Impressively, aged euglycemic mice display significant transient rise in CD4(+)CD25(-)FoxP3(+) Treg in the thymus, pancreas and draining lymph nodes. A significant difference was superior viability of effector and suppressor cells from new onset diabetics in the presence of high interleukin-2 (IL-2) concentrations in vitro as compared to cells of prediabetic mice. Overall, we found no correlation between FoxP3(+) Treg in the pancreatic lymph nodes and the inflammatory scores of individual NOD mice. CD25(-)FoxP3(+) Treg are markedly increased in the pancreatic infiltrates in late stages of inflammation, possibly an effort to counteract destructive insulitis. Considering extensive evidence that Treg in aged NOD mice are functionally sufficient, quantitative profiling evolves as an unreliable tool to assess mechanism and causes of inflammation under baseline conditions. Immune profiles are modulated by thymic output, cell migration, shedding of markers, proliferation, survival and in-situ evolution of regulatory cells.


Subject(s)
Aging/immunology , Diabetes Mellitus, Type 1/immunology , Hyperglycemia/immunology , Lymph Nodes/immunology , T-Lymphocytes, Regulatory/immunology , Aging/blood , Aging/pathology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Blood Glucose/metabolism , Cell Movement , Cells, Cultured , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/pathology , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression , Humans , Hyperglycemia/blood , Hyperglycemia/pathology , Immunophenotyping , Interleukin-2/pharmacology , Lymph Nodes/pathology , Lymphocyte Count , Mesentery/immunology , Mesentery/pathology , Mice , Mice, Inbred NOD , Pancreas/immunology , Pancreas/pathology , Severity of Illness Index , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/pathology , Thymus Gland/immunology , Thymus Gland/pathology
9.
World J Biol Psychiatry ; 15(1): 76-82, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24219803

ABSTRACT

UNLABELLED: OBJECTIVES. Disrupted in schizophrenia 1 (DISC1) is considered the most prominent candidate gene for schizophrenia. In this study, we aimed to characterize behavioural and brain biochemical traits in a mouse expressing a dominant negative DISC1mutant (DN-DISC1). METHODS: DN-DISC1 mice underwent behavioural tests to evaluate object recognition, social preference and social novelty seeking. ELISA was conducted on brain tissue to evaluate BDNF levels. Western blot was employed to measure BDNF receptor (TrkB) and cannabinoid receptor CB1. RESULTS: The mutant DISC1 mice displayed deficits in preference to social novelty while both social preference and object recognition were intact. Biochemical analysis of prefrontal cortex and hippocampus revealed a modest reduction in cortical TrkB protein levels of male mice while no differences in BDNF levels were observed. We found sex dependent differences in the expression of cannabinoid-1 receptors. CONCLUSIONS: We describe novel behavioural and biochemical abnormalities in the DN-DISC1 mouse model of schizophrenia. The data shows for the first time a possible link between DISC1 mutation and the cannabinoid system.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Nerve Tissue Proteins/genetics , Prefrontal Cortex/metabolism , Receptor, Cannabinoid, CB1/genetics , Schizophrenia/genetics , Social Behavior , Animals , Disease Models, Animal , Genes, Dominant/genetics , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice , Mice, Inbred CBA , Mice, Transgenic , Mutation , Prefrontal Cortex/pathology , Receptor, trkB/genetics , Schizophrenia/metabolism , Schizophrenia/physiopathology
10.
Exp Hematol ; 41(10): 903-11, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23743300

ABSTRACT

Infusion of large numbers of donor regulatory T cells (Tregs) is an effective approach to suppress graft-versus-host disease (GvHD). We have reported previously that enhancing the killing activity of CD25(+) Tregs by decoration with short-lived Fas-ligand (FasL) protein (killer Tregs) is effective in abrogation of autoimmunity. In this study, we assessed the therapeutic efficacy of killer Tregs in murine models of lethal GvHD. In a model in which disease-associated mortality was not prevented by infusion of naive donor Tregs (3 days after transplant) at an effector:suppressor ratio of 10:1, killer Tregs rescued 70% of the mice and improved the clinical and histologic scores. We found that both effector lymphocytes and therapeutic Tregs migrate to and proliferate in the mesenteric lymph nodes of irradiated recipients; however, only killer Tregs increased fractional apoptosis of effector lymphocytes. Although the lymphoid organs were primarily reconstituted from the bone marrow with little contribution of the infused effector and suppressor subsets, immunomodulation with FasL caused a durable rise in fractions of CD4(+)FoxP3(+) Tregs. Our findings demonstrate that a short-lived apoptotic protein increases the suppressive activity of Tregs and ameliorates GvHD severity.


Subject(s)
Fas Ligand Protein/immunology , Graft vs Host Disease/therapy , Immunomodulation , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/transplantation , Animals , Cell Movement , Cell Proliferation , Disease Models, Animal , Flow Cytometry , Graft vs Host Disease/immunology , Lymph Nodes/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/cytology
11.
Int Immunol ; 25(8): 485-94, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23657001

ABSTRACT

Treg cells endowed with enhanced killing activity through decoration with Fas-ligand (FasL) protein (killer Treg) have been effective in delay of hyperglycemia in prediabetic non-obese diabetic (NOD) mice. In this study, we assessed the therapeutic efficacy of these cells, harvested from age-matched euglycemic NOD donors, on the course of disease in new-onset diabetics. One dose of 4 × 10(6) killer Treg cells stabilized blood glucose associated with increased insulin levels in 5 of 9 mice and partially reversed the severity of islet inflammation, whereas naive Treg cells did not modulate the course of disease significantly. Killer Treg cells were shown to operate through induction of cell apoptosis within the pancreatic lymph nodes, resulting in reduced efficiency of adoptive disease transfer to NOD/SCID recipients. A second mechanism of action consisted of increased fractions of CD4(+)CD25(-)FoxP3(+) T cells in the pancreas and all lymphoid organs. Immunomodulation with FasL rather than Treg cells enhanced the expression of CD25 and FoxP3 in the thymus, suggesting a possible contribution of thymic output to prolonged stabilization of the glucose levels. Autologous Treg cells evolve as excellent vehicles for targeted delivery of FasL as an immunomodulatory protein, which delete pathogenic cells at the site of inflammation and induce systemic dominance of suppressor subsets.


Subject(s)
Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Immunomodulation , Islets of Langerhans/immunology , Islets of Langerhans/pathology , T-Lymphocytes, Regulatory/immunology , Animals , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Inbred NOD , Mice, SCID
13.
PLoS One ; 6(6): e21630, 2011.
Article in English | MEDLINE | ID: mdl-21738739

ABSTRACT

BACKGROUND: Disturbed peripheral negative regulation might contribute to evolution of autoimmune insulitis in type 1 diabetes. This study evaluates the sensitivity of naïve/effector (Teff) and regulatory T cells (Treg) to activation-induced cell death mediated by Fas cross-linking in NOD and wild-type mice. PRINCIPAL FINDINGS: Both effector (CD25(-), FoxP3(-)) and suppressor (CD25(+), FoxP3(+)) CD4(+) T cells are negatively regulated by Fas cross-linking in mixed splenocyte populations of NOD, wild type mice and FoxP3-GFP trangeneess. Proliferation rates and sensitivity to Fas cross-linking are dissociated in Treg cells: fast cycling induced by IL-2 and CD3/CD28 stimulation improve Treg resistance to Fas-ligand (FasL) in both strains. The effector and suppressor CD4(+) subsets display balanced sensitivity to negative regulation under baseline conditions, IL-2 and CD3/CD28 stimulation, indicating that stimulation does not perturb immune homeostasis in NOD mice. Effective autocrine apoptosis of diabetogenic cells was evident from delayed onset and reduced incidence of adoptive disease transfer into NOD.SCID by CD4(+)CD25(-) T cells decorated with FasL protein. Treg resistant to Fas-mediated apoptosis retain suppressive activity in vitro. The only detectable differential response was reduced Teff proliferation and upregulation of CD25 following CD3-activation in NOD mice. CONCLUSION: These data document negative regulation of effector and suppressor cells by Fas cross-linking and dissociation between sensitivity to apoptosis and proliferation in stimulated Treg. There is no evidence that perturbed AICD in NOD mice initiates or promotes autoimmune insulitis.


Subject(s)
Cell Death/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Apoptosis/drug effects , CD28 Antigens/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Fas Ligand Protein/metabolism , Flow Cytometry , Forkhead Transcription Factors/metabolism , Interleukin-2/pharmacology , Interleukin-2 Receptor alpha Subunit/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD
14.
J Autoimmun ; 37(1): 39-47, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21497486

ABSTRACT

We hypothesized that regulatory T cells (Treg) effectively target diabetogenic cells, and reinforcing their killing capacity will attenuate the course of disease. For proof of concept, Fas-ligand (FasL) protein was conjugated to CD25(+) Treg (killer Treg) to simulate the physiological mechanism of activation-induced cell death. Cytotoxic and suppressive activity of killer Treg was superior to naïve Treg in vitro. Administration of 3-4 × 10(6) Treg prevented hyperglycemia in 65% prediabetic NOD females, however only killer Treg postponed disease onset by 14 weeks. CD25(+) Treg homed to the pancreas and regional lymph nodes of prediabetic NOD females, proliferated and ectopic FasL protein induced apoptosis in CD25(-) T cells in situ. This mechanism of pathogenic cell debulking is specific to killer Treg, as FasL-coated splenocytes have no immunomodulatory effect, and only killer Treg prevent the disease in 80% of NOD.SCID recipients of effector:suppressor T cells (10:1 ratio). All immunomodulated mice displayed increased fractional expression of FoxP3 in the pancreas and draining lymph nodes, which was accompanied by CD25 only in recipients of killer Treg. A therapeutic intervention that uses the affinity of Treg to reduce the pathogenic load has long-term consequences: arrest of destructive insulitis in mice with established disease prior to ß-cell extinction.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Homeostasis/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Fas Ligand Protein/immunology , Female , Immunomodulation , Inflammation/immunology , Inflammation/pathology , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Mice , Mice, Inbred NOD , Mice, SCID
15.
J Autoimmun ; 35(2): 145-52, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20638242

ABSTRACT

Mounting evidence indicates that immunosuppressive therapy and autologous bone marrow transplantation are relatively inefficient approaches to treat autoimmune diabetes. In this study we assessed the impact of immunosuppression on inflammatory insulitis in NOD mice, and the effect of radiation on immunomodulation mediated by adoptive transfer of various cell subsets. Sublethal radiation of NOD females at the age of 14 weeks (onset of hyperglycemia) delayed the onset of hyperglycemia, however two thirds of the mice became diabetic. Adoptive transfer of splenocytes into irradiated NON and NOD mice precipitated disease onset despite increased contents of CD25(+)FoxP3(+) T cells in the pancreas and regional lymphatics. Similar phenotypic changes were observed when CD25(+) T cells were infused after radiation, which also delayed disease onset without affecting its incidence. Importantly, irradiation increased the susceptibility to diabetes in NOD and NON mice (71-84%) as compared to immunomodulation with splenocytes and CD25(+) T cells in naïve recipients (44-50%). Although irradiation had significant and durable influence on pancreatic infiltrates and the fractions of functional CD25(+)FoxP3(+) Treg cells were elevated by adoptive cell transfer, this approach conferred no protection from disease progression. Irradiation was ineffective both in debulking of pathogenic clones and in restoring immune homeostasis, and the consequent homeostatic expansion evolves as an unfavorable factor in attempts to restore self-tolerance and might even provoke uncontrolled proliferation of pathogenic clones. The obstacles imposed by immunosuppression on abrogation of autoimmune insulitis require replacement of non-specific immunosuppressive therapy by selective immunomodulation that does not cause lymphopenia.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , Immunotherapy , Radiotherapy , T-Lymphocytes, Regulatory/metabolism , Adoptive Transfer , Animals , Autoimmunity/radiation effects , Cell Count , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/physiopathology , Disease Susceptibility/etiology , Female , Forkhead Transcription Factors/biosynthesis , Hyperglycemia , Immunomodulation/radiation effects , Interleukin-2 Receptor alpha Subunit/biosynthesis , Mice , Mice, Inbred NOD , Pancreas/pathology , Radiotherapy/adverse effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , T-Lymphocytes, Regulatory/radiation effects
16.
PLoS One ; 5(12): e15684, 2010 Dec 31.
Article in English | MEDLINE | ID: mdl-21209873

ABSTRACT

BACKGROUND: Regulatory T cells (Treg) play a significant role in immune homeostasis and self-tolerance. Excessive sensitivity of isolated Treg to apoptosis has been demonstrated in NOD mice and humans suffering of type 1 diabetes, suggesting a possible role in the immune dysfunction that underlies autoimmune insulitis. In this study the sensitivity to apoptosis was measured in T cells from new onset diabetic NOD females, comparing purified subsets to mixed cultures. PRINCIPAL FINDINGS: Apoptotic cells are short lived in vivo and death occurs primarily during isolation, manipulation and culture. Excessive susceptibility of CD25(+) T cells to spontaneous apoptosis is characteristic of isolated subsets, however disappears when death is measured in mixed splenocyte cultures. In variance, CD25(-) T cells display balanced sensitivity to apoptosis under both conditions. The isolation procedure removes soluble factors, IL-2 playing a significant role in sustaining Treg viability. In addition, pro- and anti-apoptotic signals are transduced by cell-to-cell interactions: CD3 and CD28 protect CD25(+) T cells from apoptosis, and in parallel sensitize naïve effector cells to apoptosis. Treg viability is modulated both by other T cells and other subsets within mixed splenocyte cultures. Variations in sensitivity to apoptosis are often hindered by fast proliferation of viable cells, therefore cycling rates are mandatory to adequate interpretation of cell death assays. CONCLUSIONS: The sensitivity of purified Treg to apoptosis is dominated by cytokine deprivation and absence of cell-to-cell interactions, and deviate significantly from measurements in mixed populations. Balanced sensitivity of naïve/effector and regulatory T cells to apoptosis in NOD mice argues against the concept that differential susceptibility affects disease evolution and progression.


Subject(s)
Apoptosis , CD4-Positive T-Lymphocytes/cytology , Cytokines/metabolism , Animals , CD28 Antigens/biosynthesis , CD3 Complex/biosynthesis , Cell Death , Diabetes Mellitus, Experimental/metabolism , Female , Homeostasis , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Inbred NOD
17.
Ann N Y Acad Sci ; 1173: 791-7, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19758230

ABSTRACT

Targeted depletion of immune cells expressing the interleukin-2 (IL-2) receptor can exacerbate inflammatory bowel disease (IBD) through elimination of regulatory T (Treg) cells, or ameliorate its course by depletion of cytotoxic cells. To answer this question we used a fusion protein composed of IL-2 and caspase-3 (IL2-cas) in an experimental model of DSS-induced toxic colitis. In a preventive setting, co-administration of DSS with a daily therapeutic dose of IL2-cas for seven days improved all disease parameters. Although CD4(+)CD25(+) T cells were depleted in the mesenteric lymph nodes, a fractional increase in CD4(+)FoxP3(+) T cells was observed in the spleen. Likewise, IL2-cas therapy improved the outcome of established disease in a chronic model of colitis. These data demonstrate that therapies that use IL-2 as a targeting moiety exert a protective effect over the colon under conditions of inflammation. The efficacy of IL-2-targeted therapy is attributed to reduced activity of reactive T cells, which ameliorates the secondary inflammatory infiltration. IL2-cas evolves as a potential therapeutic tool in IBD.


Subject(s)
Caspase 3/metabolism , Colitis/prevention & control , Interleukin-2/metabolism , Recombinant Fusion Proteins/pharmacology , Animals , Body Weight/drug effects , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Caspase 3/genetics , Colitis/chemically induced , Dextran Sulfate , Forkhead Transcription Factors/metabolism , Injections, Intravenous , Interleukin-2/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/metabolism , Spleen/cytology , Spleen/drug effects , Spleen/metabolism , Time Factors , Treatment Outcome
18.
Eur J Immunol ; 39(10): 2850-64, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19735074

ABSTRACT

Pathogenic lymphocytes in the enteric wall of inflammatory bowel disease patients display various abnormalities, including reduced sensitivity to apoptosis. We evaluated a therapeutic approach to elimination of cytotoxic cells, using two IL-2 fusion proteins, a diphtheria toxin (IL2-DT) and a caspase-3 (IL2-cas) conjugate. In models of acute (dextran sodium sulfate and trinitrobenzene sulfonic acid) and chronic (dextran sodium sulfate) toxic colitis, therapeutic doses of the fusion proteins improved survival and prevented colon shortening. While both chimeric proteins eradicated CD4(+)CD25(+)Foxp3(+) T cells in mesenteric LN, IL2-DT caused severe lymphopenia. In contrast, IL2-cas was equally protective and increased fractional expression of Foxp3. Similar effects of the fusion proteins were observed in healthy mice: IL2-DT caused lymphopenia and IL2-cas increased fractional expression of FoxP3. The fusion proteins induced apoptosis in CD25(+) T cells in vitro, with lower toxicity of IL2-cas to Foxp3(+) T cells. These data infer that targeted depletion of cells expressing the IL-2 receptor has therapeutic potential in models of inflammatory colitis, despite depletion of CD25(+) Treg. The IL2-cas fusion protein is particularly relevant to inflammatory bowel disease, as direct internalization of toxic moieties overcomes multiple pathways of resistance to apoptosis of colitogenic T cells.


Subject(s)
Caspase 3/therapeutic use , Diphtheria Toxin/therapeutic use , Drug Delivery Systems/methods , Inflammatory Bowel Diseases/drug therapy , Receptors, Interleukin-2/metabolism , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes, Regulatory/drug effects , Animals , Apoptosis/drug effects , Apoptosis/immunology , Body Weight/drug effects , Caspase 3/administration & dosage , Caspase 3/genetics , Caspase 3/pharmacology , Cell Proliferation/drug effects , Colon/drug effects , Colon/pathology , Dextran Sulfate/administration & dosage , Dextran Sulfate/pharmacology , Diphtheria Toxin/administration & dosage , Diphtheria Toxin/genetics , Diphtheria Toxin/pharmacology , Forkhead Transcription Factors/metabolism , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Interleukin-2/genetics , Interleukin-2/metabolism , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/pathology , Male , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Spleen/drug effects , Spleen/immunology , Spleen/pathology , Survival Analysis , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Trinitrobenzenesulfonic Acid/administration & dosage , Trinitrobenzenesulfonic Acid/pharmacology
19.
J Autoimmun ; 33(2): 83-91, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19632089

ABSTRACT

The relative efficiencies of allogeneic and syngeneic bone marrow transplantation and the threshold levels of donor chimerism required to control autoimmune insulitis were evaluated in prediabetic NOD mice. Male and female NOD mice were conditioned by radiation and grafted with bone marrow cells from allogeneic and syngeneic sex-mismatched donors. Establishment of full allogeneic chimerism in peripheral blood reversed insulitis and restored glucose tolerance despite persistence of residual host immune cells. By contrast, sublethal total body irradiation (with or without syngeneic transplant) reduced the incidence and delayed the onset of diabetes. The latter pattern was also seen in mice that rejected the bone marrow allografts. Low levels of stable allogeneic hematopoietic chimerism (>1%) were sufficient to prevent the evolution of diabetes following allogeneic transplantation. The data indicate that immunomodulation attained at low levels of allogeneic, but not syngeneic, hematopoietic chimerism is effective in resolution of islet inflammation at even relatively late stages in the evolution of the prediabetic state in a preclinical model. However, our data question the efficacy and rationale behind syngeneic (autologous-like) immuno-hematopoietic reconstitution in type 1 diabetes.


Subject(s)
Bone Marrow Transplantation , Diabetes Mellitus, Type 1/prevention & control , Prediabetic State/surgery , Age Factors , Animals , Autoimmunity/immunology , Chimerism , Female , Inflammation/surgery , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Transplantation Conditioning , Transplantation, Homologous , Transplantation, Isogeneic
20.
Immunol Cell Biol ; 86(8): 650-8, 2008.
Article in English | MEDLINE | ID: mdl-18794907

ABSTRACT

Activation-induced cell death is a general mechanism of immune homeostasis through negative regulation of clonal expansion of activated immune cells. This mechanism is involved in the maintenance of self- and transplant tolerance through polarization of the immune responses. The Fas/Fas-ligand interaction is a major common executioner of apoptosis in lymphocytes, with a dual role in regulatory T cell (Treg) function: Treg cell homeostasis and Treg cell-mediated suppression. Sensitivity to apoptosis and the patterns of Treg-cell death are of outmost importance in immune homeostasis that affects the equilibrium between cytolytic and suppressor forces in activation and termination of immune activity. Naive innate (naturally occurring) Treg cells present variable sensitivities to apoptosis, related to their turnover rates in tissue under steady state conditions. Following activation, Treg cells are less sensitive to apoptosis than cytotoxic effector subsets. Their susceptibility to apoptosis is influenced by cytokines within the inflammatory environment (primarily interleukin-2), the mode of antigenic stimulation and the proliferation rates. Here, we attempt to resolve some controversies surrounding the sensitivity of Treg cells to apoptosis under various experimental conditions, to delineate the function of cell death in regulation of immunity.


Subject(s)
Apoptosis , Cytokines/immunology , Homeostasis , T-Lymphocytes, Regulatory/immunology , Animals , Antineoplastic Agents/pharmacology , Cyclophosphamide/pharmacology , Cytokines/metabolism , Fas Ligand Protein/metabolism , Fas-Associated Death Domain Protein/metabolism , Humans , Immunosuppressive Agents/pharmacology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Vidarabine/analogs & derivatives , Vidarabine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL