Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Immunology ; 167(3): 303-313, 2022 11.
Article in English | MEDLINE | ID: mdl-35752961

ABSTRACT

Autoreactive T cells with the phenotype and function of different memory subsets are present in patients who developed type 1 diabetes (TID). According to the progressive differentiation model, memory subsets generate from naïve precursors in a linear and unidirectional path depending on the strength and quality of stimulatory signals. By observing human naïve T cells in contact with GAD65 loaded autologous dendritic cells, we observed that approximately 10% of cells divided with the plane of cell division parallel to the one of the immune synapse, causing phenotypic asymmetries in the proximal and distal daughter T cells. After the first T cell division, proximal and distal daughter T cells showed different phenotype, metabolic signature and commitment to differentiate towards long-lived memory T cells or T cells with effector function. Subjects with or without T1D showed a similar frequency of asymmetric T cell division (ATCD) for autoantigens and recall antigens specific T cells, however the frequency of ATCD is significantly increased in autoreactive T cells in patients with T1D when IL-7 was added to the culture. An increased upregulation of GLUT1 in response to IL-7 in patients with T1D was related to the rate of ATCD. Our results showed that ATCD is associated with an early divergence in the differentiation fate of naïve T cells specific for GAD65 during first antigen encounter.


Subject(s)
Diabetes Mellitus, Type 1 , Autoantigens , Cell Differentiation , Cell Division , Glucose Transporter Type 1/metabolism , Humans , Immunologic Memory , Interleukin-7/metabolism , Memory T Cells , T-Lymphocyte Subsets
2.
Cancers (Basel) ; 12(10)2020 Oct 12.
Article in English | MEDLINE | ID: mdl-33053779

ABSTRACT

The orchestration of T cell responses is intimately linked to the execution of metabolic processes, both in homeostasis and disease. In cancer tissues, metabolic alterations that characterize malignant transformation profoundly affect the composition of the immune microenvironment and the accomplishment of an effective anti-tumor response. The growing understanding of the metabolic regulation of immune cell function has shed light on the possibility to manipulate metabolic pathways as a strategy to improve T cell function in cancer. Among others, glucose metabolism through the glycolytic pathway is central in shaping T cell responses and emerges as an ideal target to improve cancer immunotherapy. However, metabolic manipulation requires a deep level of control over side-effects and development of biomarkers of response. Here, we summarize the metabolic control of T cell function and focus on the implications of metabolic manipulation for the design of immunotherapeutic strategies. Integrating our understanding of T cell function and metabolism will hopefully foster the forthcoming development of more effective immunotherapeutic strategies.

4.
Cancer Immunol Res ; 8(4): 493-505, 2020 04.
Article in English | MEDLINE | ID: mdl-32019781

ABSTRACT

Better understanding of pancreatic diseases, including pancreatic ductal adenocarcinoma (PDAC), is an urgent medical need, with little advances in preoperative differential diagnosis, preventing rational selection of therapeutic strategies. The clinical management of pancreatic cancer patients would benefit from the identification of variables distinctively associated with the multiplicity of pancreatic disorders. We investigated, by 1H nuclear magnetic resonance, the metabolomic fingerprint of pancreatic juice (the biofluid that collects pancreatic products) in 40 patients with different pancreatic diseases. Metabolic variables discriminated PDAC from other less aggressive pancreatic diseases and identified metabolic clusters of patients with distinct clinical behaviors. PDAC specimens were overtly glycolytic, with significant accumulation of lactate, which was probed as a disease-specific variable in pancreatic juice from a larger cohort of 106 patients. In human PDAC sections, high expression of the glucose transporter GLUT-1 correlated with tumor grade and a higher density of PD-1+ T cells, suggesting their accumulation in glycolytic tumors. In a preclinical model, PD-1+ CD8 tumor-infiltrating lymphocytes differentially infiltrated PDAC tumors obtained from cell lines with different metabolic consumption, and tumors metabolically rewired by knocking down the phosphofructokinase (Pfkm) gene displayed a decrease in PD-1+ cell infiltration. Collectively, we introduced pancreatic juice as a valuable source of metabolic variables that could contribute to differential diagnosis. The correlation of metabolic markers with immune infiltration suggests that upfront evaluation of the metabolic profile of PDAC patients could foster the introduction of immunotherapeutic approaches for pancreatic cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Metabolome , Pancreatic Juice/metabolism , Pancreatic Neoplasms/pathology , Programmed Cell Death 1 Receptor/metabolism , Aged , Animals , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/metabolism , Cells, Cultured , Coculture Techniques , Female , Glucose Transporter Type 1/metabolism , Humans , Leukocytes, Mononuclear/immunology , Male , Mice , Mice, Transgenic , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Programmed Cell Death 1 Receptor/immunology , Survival Rate
5.
Int J Mol Sci ; 20(19)2019 Oct 08.
Article in English | MEDLINE | ID: mdl-31597342

ABSTRACT

An increasing body of evidence indicates that bio-energetic metabolism of T cells can be manipulated to control T cell responses. This potentially finds a field of application in the control of the T cell responses in autoimmune diseases, including in type 1 diabetes (T1D). Of the possible metabolic targets, Glut1 gained considerable interest because of its pivotal role in glucose uptake to fuel glycolysis in activated T cells, and the recent development of a novel class of small molecules that act as selective inhibitor of Glut1. We believe we can foresee a possible application of pharmacological Glut1 blockade approach to control autoreactive T cells that destroy insulin producing beta cells. However, Glut1 is expressed in a broad range of cells in the body and off-target and side effect are possible complications. Moreover, the duration of the treatment and the age of patients are critical aspects that need to be addressed to reduce toxicity. In this paper, we will review recent literature to determine whether it is possible to design a pharmacological Glut1 blocking strategy and how to apply this to autoimmunity in T1D.


Subject(s)
Autoimmunity , Glucose Transporter Type 1/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Autoimmune Diseases/diagnosis , Autoimmune Diseases/drug therapy , Autoimmune Diseases/etiology , Autoimmune Diseases/metabolism , Autoimmunity/drug effects , Energy Metabolism/drug effects , Gene Expression Regulation , Glucose Transporter Type 1/antagonists & inhibitors , Glucose Transporter Type 1/chemistry , Glucose Transporter Type 1/genetics , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Molecular Targeted Therapy , Signal Transduction , Structure-Activity Relationship , T-Lymphocytes/drug effects
6.
Diabetes ; 67(5): 936-945, 2018 05.
Article in English | MEDLINE | ID: mdl-29506985

ABSTRACT

Stem memory T cells (Tscm) constitute the earliest developmental stage of memory T cells, displaying stem cell-like properties, such as self-renewal capacity. Their superior immune reconstitution potential has sparked interest in cancer immune therapy, vaccine development, and immune reconstitution, whereas their role in autoimmunity is largely unexplored. Here we show that autoreactive CD8+ Tscm specific for ß-cell antigens GAD65, insulin, and IGRP are present in patients with type 1 diabetes (T1D). In vitro, the generation of autoreactive Tscm from naive precursors required the presence of the homeostatic cytokine interleukin-7 (IL-7). IL-7 promotes glucose uptake via overexpression of GLUT1 and upregulation of the glycolytic enzyme hexokinase 2. Even though metabolism depends on glucose uptake, the subsequent oxidation of pyruvate in the mitochondria was necessary for Tscm generation from naive precursors. In patients with T1D, high expression of GLUT1 was a hallmark of circulating Tscm, and targeting glucose uptake via GLUT1 using the selective inhibitor WZB117 resulted in inhibition of Tscm generation and expansion. Our results suggest that autoreactive Tscm are present in patients with T1D and can be selectively targeted by inhibition of glucose metabolism.


Subject(s)
Autoimmunity/immunology , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Lymphoid Progenitor Cells/immunology , T-Lymphocyte Subsets/immunology , Adolescent , Adult , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Child , Diabetes Mellitus, Type 1/metabolism , Female , Glucose/metabolism , Glucose Transporter Type 1/metabolism , Glucose-6-Phosphatase/immunology , Glutamate Decarboxylase/immunology , Hexokinase/metabolism , Humans , Hydroxybenzoates/pharmacology , Immunologic Memory/immunology , In Vitro Techniques , Insulin/immunology , Interleukin-7/immunology , Lymphopoiesis/drug effects , Male , Middle Aged , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/metabolism , Up-Regulation
7.
Cytokine Growth Factor Rev ; 36: 107-116, 2017 08.
Article in English | MEDLINE | ID: mdl-28690108

ABSTRACT

Cytotoxic T lymphocytes (cytotoxic T cells, CTLs) are an immune effector cell population that can mediate specific immune responses against cancer. Based on this concept, tumor immunotherapy protocols have been developed using adoptive transfer of in vitro-expanded autologous T cells that can kill cancer cells. However, fully functional adoptive T cell therapies (ACT) are hampered by the inability to guarantee that all transferred T cells manage to reach the tumor sites and make contact with cancer cells. The lack of tumor homing of T cells may be caused by a variety of reasons. Stromal architecture and biological features of the tumor microenvironment may act as barriers to T cell migration. A mismatch between the chemokines released by the tumor or tumor stroma and the chemokine receptors expressed on the transferred T cells may also impede T cell homing. The identification of mechanisms responsible for cancer stroma remodeling is helping to overcome the barriers of access to tumors, via novel therapeutic strategies targeting tumor-stroma interactions. Simultaneously, recent studies have demonstrated ways through which virally-transduced CTLs can be made to express suitable chemokine receptors so as to enhance ACT, by improving CTL homing into the tumor. Here we review the most important findings related to T cell trafficking to the tumor, highlighting contributions that have led to promising improvements in the available T cell therapy strategies. We discuss new possible combinatorial strategies aimed to overcome chemokine mismatch, physical and biological barriers and immunosuppression, so as to achieve more effective ACT therapies.


Subject(s)
Chemokines/immunology , Immunotherapy, Adoptive/methods , Immunotherapy/methods , Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Movement , Humans , Mice , Neoplasms/immunology , Receptors, Chemokine/immunology , Tumor Microenvironment/immunology
8.
Nat Commun ; 8: 14680, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28262700

ABSTRACT

Heart failure (HF) is a leading cause of mortality. Inflammation is implicated in HF, yet clinical trials targeting pro-inflammatory cytokines in HF were unsuccessful, possibly due to redundant functions of individual cytokines. Searching for better cardiac inflammation targets, here we link T cells with HF development in a mouse model of pathological cardiac hypertrophy and in human HF patients. T cell costimulation blockade, through FDA-approved rheumatoid arthritis drug abatacept, leads to highly significant delay in progression and decreased severity of cardiac dysfunction in the mouse HF model. The therapeutic effect occurs via inhibition of activation and cardiac infiltration of T cells and macrophages, leading to reduced cardiomyocyte death. Abatacept treatment also induces production of anti-inflammatory cytokine interleukin-10 (IL-10). IL-10-deficient mice are refractive to treatment, while protection could be rescued by transfer of IL-10-sufficient B cells. These results suggest that T cell costimulation blockade might be therapeutically exploited to treat HF.


Subject(s)
Cardiomegaly/metabolism , Heart Failure/metabolism , Macrophages/metabolism , T-Lymphocytes/metabolism , Abatacept/pharmacology , Animals , Animals, Newborn , Cardiomegaly/genetics , Cardiomegaly/prevention & control , Cells, Cultured , Heart Failure/genetics , Heart Failure/prevention & control , Humans , Immunosuppressive Agents/pharmacology , Interleukin-10/genetics , Interleukin-10/metabolism , Macrophages/drug effects , Male , Mice, Inbred C57BL , Mice, Knockout , Pressure , T-Lymphocytes/drug effects
9.
Transplantation ; 100(9): 1853-61, 2016 09.
Article in English | MEDLINE | ID: mdl-27306531

ABSTRACT

BACKGROUND: The maintenance or expansion of regulatory T (Treg) cells has a fundamental role in the achievement of immunological tolerance after transplantation. Here we aimed to determine mechanisms of human Treg cell depletion and reconstitution after anti-CD25 monoclonal antibody (mAb) treatment. METHODS: Seventeen patients with type 1 diabetes who received pancreatic islet transplantation and anti-CD25 mAb as induction therapy were studied. RESULTS: We observed an almost complete depletion of Treg cells after injection of anti-CD25 mAb. The kinetic of Treg cell depletion did not parallel the disappearance of CD25+ T cells as CD25 is also rapidly downregulated and internalized. Regulatory T cell reconstitution is completed within 6 months posttransplantation and appeared to be driven by IL-7-mediated homeostatic T cell proliferation. Anti-CD25 mAb treatment sensitizes Treg cell to the biological effect of IL-7, possibly rendering more common γc-chain available to interact with CD127. Homeostatic Treg cell proliferation is resistant to the inhibitory effect of rapamycin and FK506 but can be blocked by the presence of mycophenolate mofetil. CONCLUSIONS: Our data suggest that a compensatory mechanism of IL-7-mediated homeostatic proliferation can restore the inhibitory network of Treg cell after anti-CD25 induction therapy in islet allotransplantation.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Cell Proliferation/drug effects , Diabetes Mellitus, Type 1/surgery , Forkhead Transcription Factors/immunology , Immunosuppressive Agents/therapeutic use , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-7/pharmacology , Islets of Langerhans Transplantation , Lymphocyte Depletion/methods , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes, Regulatory/drug effects , Adult , Allografts , Antibodies, Monoclonal/adverse effects , Basiliximab , Cells, Cultured , Diabetes Mellitus, Type 1/diagnosis , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , Forkhead Transcription Factors/blood , Humans , Immunologic Memory/drug effects , Immunosuppressive Agents/adverse effects , Interleukin Receptor Common gamma Subunit/immunology , Interleukin-2 Receptor alpha Subunit/blood , Interleukin-7 Receptor alpha Subunit/immunology , Islets of Langerhans Transplantation/adverse effects , Male , Middle Aged , Mycophenolic Acid/therapeutic use , Recombinant Fusion Proteins/adverse effects , Signal Transduction/drug effects , Sirolimus/therapeutic use , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Tacrolimus/therapeutic use , Time Factors , Treatment Outcome
10.
Oncotarget ; 7(28): 43010-43026, 2016 Jul 12.
Article in English | MEDLINE | ID: mdl-27177227

ABSTRACT

In recent years, tumor Adoptive Cell Therapy (ACT), using administration of ex vivo-enhanced T cells from the cancer patient, has become a promising therapeutic strategy. However, efficient homing of the anti-tumoral T cells to the tumor or metastatic site still remains a substantial hurdle. Yet the tumor site itself attracts both tumor-promoting and anti-tumoral immune cell populations through the secretion of chemokines. We attempted to identify these chemokines in a model of spontaneous metastasis, in order to "hijack" their function by expressing matching chemokine receptors on the cytotoxic T cells used in ACT, thus allowing us to enhance the recruitment of these therapeutic cells. Here we show that this enabled the modified T cells to preferentially home into spontaneous lymph node metastases in the TRAMP model, as well as in an inducible tumor model, E.G7-OVA. Due to the improved homing, the modified CD8+ T cells displayed an enhanced in vivo protective effect, as seen by a significant delay in E.G7-OVA tumor growth. These results offer a proof of principle for the tailored application of chemokine receptor modification as a means of improving T cell homing to the target tumor, thus enhancing ACT efficacy. Surprisingly, we also uncover that the formation of the peri-tumoral fibrotic capsule, which has been shown to impede T cell access to tumor, is partially dependent on host T cell presence. This finding, which would be impossible to observe in immunodeficient model studies, highlights possible conflicting roles that T cells may play in a therapeutic context.


Subject(s)
Immunotherapy, Adoptive/methods , Neoplasms, Experimental/therapy , Receptors, Chemokine/immunology , T-Lymphocytes/immunology , Animals , Cell Line, Tumor , Cells, Cultured , Cytotoxicity, Immunologic/immunology , Gene Expression Regulation, Neoplastic/immunology , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism
11.
Curr Diab Rep ; 16(5): 40, 2016 May.
Article in English | MEDLINE | ID: mdl-26983628

ABSTRACT

Immunomodulation of the autoreactive T cell response is considered a major strategy to control beta-cell autoimmunity, both in the natural history of type 1 diabetes and in islet transplantation, which can be affected by autoimmunity recurrence. So far, these strategies have had modest results, prompting efforts to define novel cellular and molecular targets to control autoreactive T cell expansion and activation. Novel findings highlighted the important role of the homeostatic cytokine interleukin-7 in inducing proliferation and differentiation of autoreactive T cell clones that causes beta-cell autoimmunity. In this review, we discuss recent evidences and novel findings on the role of IL-7 mediated homeostatic T cell proliferation in the process of beta-cell destruction and evidences of how targeting IL-7 and its receptor could be an innovative and effective strategy to control beta-cell autoimmunity.


Subject(s)
Autoimmunity , Cell Proliferation , Homeostasis , Insulin-Secreting Cells/immunology , T-Lymphocytes/immunology , Animals , Humans , Interleukin-7/immunology , T-Lymphocytes/cytology
12.
Curr Diabetes Rev ; 11(3): 135-43, 2015.
Article in English | MEDLINE | ID: mdl-25777058

ABSTRACT

Type 1 diabetes (T1D) is caused by the chronic autoimmune destruction of insulin producing beta cells. Beta cell replacement therapy through whole pancreas or islet transplantation is a therapeutic option for patients in which a stable glucose control is not achievable with exogenous insulin therapy. Long-term insulin independence is, however, hampered by the recipient immune response that includes activation of inflammatory pathways and specific allo- and autoimmunity. The identification and monitoring of soluble and cellular biomarkers are of critical relevance for the prediction of graft damage, for the evaluation of responses to immune-modulating therapy, and for target pathways identification to generate novel drugs or therapeutic approaches. The final objective of immune monitoring is to find ways to improve the outcome of pancreas and islet transplantation. In this review, we discuss the available tools to monitor the innate, humoral and cellular responses after islet and pancreas transplantation, and the most relevant findings generated by these measurements.


Subject(s)
Diabetes Mellitus, Type 1/surgery , Graft Rejection/immunology , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Inflammation/immunology , Islets of Langerhans Transplantation/immunology , Pancreas Transplantation , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...