Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Proteins ; 89(2): 141-148, 2021 02.
Article in English | MEDLINE | ID: mdl-32862461

ABSTRACT

Activation of T cells triggers the expression of regulatory molecules like the programmed cell death 1 (PD1) protein. The association of PD1 with the natural ligands PDL1 and PDL2 induces an inhibitory signal that prevents T cells from proliferating and exerting effector functions. However, little is known about how the binding of the ligands induce the PD1 inhibitory signal over T cells effector functions. Here, we explore the dynamics of PD1 free, and in complex with different PDL1 variants as well as the therapeutic antibodies nivolumab and pembrolizumab in order to assess the conformational changes in PD1 related to the signaling process. Our simulations suggest a pre-conformational selection mechanism for the binding of the different PDL1 variants, while an induced-fit model fits better for the molecular recognition process of the therapeutic antibodies. A deep analysis of the changes on PD1 movement upon the binding to different ligands revealed that as larger is the difference in the conformation adopted by loop C'D with respect to the complex with PDL1 is higher the ligand ability to reduce the PD1 inhibitory signaling. This behavior suggests that targeting specific conformations of this loop can be useful for designing therapies able to recover T cells effector functions.


Subject(s)
Antibodies, Monoclonal, Humanized/chemistry , B7-H1 Antigen/chemistry , Nivolumab/chemistry , Programmed Cell Death 1 Receptor/chemistry , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/metabolism , Antineoplastic Agents, Immunological , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Binding Sites , Gene Expression , Humans , Ligands , Molecular Dynamics Simulation , Nivolumab/immunology , Nivolumab/metabolism , Principal Component Analysis , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Signal Transduction
2.
J Theor Biol ; 487: 110113, 2020 02 21.
Article in English | MEDLINE | ID: mdl-31830463

ABSTRACT

IL-1 system is involved in the induction and maintenance of chronic inflammation associated with several autoimmune diseases and cancer, mainly due to its capacity to promote the secretion of inflammatory mediators. For this reason, several intracellular and extracellular mechanisms for this system have been fixed during the evolution. In spite of the large description of molecular interactions between IL-1 ligands and receptors, little is known about the relevance and limits of the extracellular regulatory mechanims in different scenarios. To tackle this problem, we developed and calibrated a mathematical model including all the known interactions between IL-1 ligands and IL-1Rs and calibrate it with experimental data of IL-1 binding to different cells. The model predicts that, independently on the IL-1Rs expression, IL-1α has more ability than IL-1ß to induce IL-1 signaling, which suggests that both ligands can be equally relevant for the IL-1 related inflammation. On the other hand, at the cell level, IL-1 signaling is mainly controlled by IL-1R1 and IL-1R3 and not by IL-1R2. Moreover, the soluble form of IL-1R1 and IL-1RA have the highest capacity to prevent IL-1α while IL-1R2 and IL-1R1 and IL-1RA have a similar capacity to prevent IL-1ß signaling. The soluble IL-1R3 has the lowest capacity to prevent IL-1 signaling and preferentially inhibits cells with low number of IL-1R3. In general, model predictions suggest several ways in which IL-1 controlling system may fail, developing IL-1 related inflammation.


Subject(s)
Autoimmune Diseases , Inflammation , Humans , Signal Transduction
3.
Semin Oncol ; 45(1-2): 95-104, 2018 01.
Article in English | MEDLINE | ID: mdl-30318089

ABSTRACT

High-dose IL2, first approved in 1992, has been used in the treatment of advanced renal cell carcinoma and melanoma. In these indications, IL2 induces long lasting objective responses in 5% to 20% of patients. However, toxicity and the unexpected expansion of regulatory T cells (Tregs) have limited its practical use and therapeutic impact, respectively. At the Center of Molecular Immunology in Havana, Cuba, a project was launched in 2005 to rationally design IL2 muteins that could be deployed in the therapy of cancer. The basic goal was to uncouple the pleiotropic effect of IL2 on different immune T cells, to obtain a mutein with a therapeutic index that was better than that achieved with wild type (wt) IL2. Using a combination of computational and experimental biology approaches, we predicted and developed two novel IL2 muteins with therapeutic potential. The first, designated no-alpha mutein, is an agonist of IL2R signaling with a reduced ability to expand Treg in vivo. In mice, the no-alpha mutein IL2 has higher antitumor activity and lower toxicity than wt IL2. It represents a potential best-in-class drug that has begun phase I/II clinical trials in solid tumors. The second, designated no-gamma mutein, is an antagonist of IL2R signaling, with some preferential affinity for Tregs. This mutein has antitumor activity in mice that likely derives from its ability to reduce Treg accumulation in vivo. It represents a first-in-class drug that offers a novel strategy to inhibit Treg activity in vivo.


Subject(s)
Computational Biology/methods , Disease Models, Animal , Interleukin-2/therapeutic use , Neoplasms/drug therapy , Animals , Humans , Interleukin-2/genetics , Interleukin-2/immunology , Mutation , Neoplasms/immunology , Receptors, Interleukin-2/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
4.
J Immunol ; 200(10): 3475-3484, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29618524

ABSTRACT

IL-2 is critical for peripheral tolerance mediated by regulatory T (Treg) cells, which represent an obstacle for effective cancer immunotherapy. Although IL-2 is important for effector (E) T cell function, it has been hypothesized that therapies blocking IL-2 signals weaken Treg cell activity, promoting immune responses. This hypothesis has been partially tested using anti-IL-2 or anti-IL-2R Abs with antitumor effects that cannot be exclusively attributed to lack of IL-2 signaling in vivo. In this work, we pursued an alternative strategy to block IL-2 signaling in vivo, taking advantage of the trimeric structure of the IL-2R. We designed an IL-2 mutant that conserves the capacity to bind to the αß-chains of the IL-2R but not to the γc-chain, thus having a reduced signaling capacity. We show our IL-2 mutein inhibits IL-2 Treg cell-dependent differentiation and expansion. Moreover, treatment with IL-2 mutein reduces Treg cell numbers and impairs tumor growth in mice. A mathematical model was used to better understand the effect of the mutein on Treg and E T cells, suggesting suitable strategies to improve its design. Our results show that it is enough to transiently inhibit IL-2 signaling to bias E and Treg cell balance in vivo toward immunity.


Subject(s)
Cell Proliferation/drug effects , Interleukin-2/antagonists & inhibitors , Lymphokines/pharmacology , Neoplasms/therapy , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/drug effects , Animals , Cell Differentiation/drug effects , Cell Line , Cell Line, Tumor , Female , Humans , Immunotherapy/methods , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/metabolism , Peripheral Tolerance/drug effects
5.
Article in English | MEDLINE | ID: mdl-29507589

ABSTRACT

Gastric ulcers are a worldwide health problem and their poor healing is one of the most important causes for their recurrence. We have previously reported the remarkable gastroprotective and anti-Helicobacter pylori activities of the methanolic extract (CpMet) of Cyrtocarpa procera bark. This work investigates, in a murine model, the CpMet gastroprotective mechanism and establishes its preclinical efficacy in the resolution of ethanol-induced gastric ulcers. The results showed that the gastroprotective activity of CpMet is mainly associated with endogenous NO and prostaglandins, followed by sulfhydryl groups and KATP channels. Furthermore, CpMet (300 mg/kg, twice a day) orally administered during 20 consecutive days promoted an ulcer area reduction of 62.65% at the 20th day of the treatment. The effect was confirmed macroscopically by the alleviation of gastric mucosal erosions and microscopically by an increase in mucin content and a reduction in the inflammatory infiltration at the site of the ulcer. No clinical symptoms or signs of toxicity were observed in the treated animals. The results indicate the safety and efficacy of CpMet in promoting high quality of ulcer healing by different mechanisms, but mostly through cytoprotective and anti-inflammatory effects, making it a promising phytodrug for ulcer treatment.

6.
PLoS One ; 11(5): e0155684, 2016.
Article in English | MEDLINE | ID: mdl-27195783

ABSTRACT

Interleukin-2 (IL2) is a growth factor for several immune cells and its function depends on its binding to IL2Rs in the cell membrane. The most accepted model for the assembling of IL2-IL2R complexes in the cell membrane is the Affinity Conversion Model (ACM). This model postulates that IL2R receptor association is sequential and dependent on ligand binding. Most likely free IL2 binds first to IL2Rα, and then this complex binds to IL2Rß, and finally to IL2Rγ (γc). However, in previous mathematical models representing this process, the binding of γc has not been taken into account. In this work, the quantitative contribution of the number of IL2Rγ chain to the IL2-IL2R apparent binding affinity and signaling is studied. A mathematical model of the affinity conversion process including the γ chain in the dynamic, has been formulated. The model was calibrated by fitting it to experimental data, specifically, Scatchard plots obtained using human cell lines. This paper demonstrates how the model correctly explains available experimental observations. It was estimated, for the first time, the value of the kinetic coefficients of IL2-IL2R complexes interaction in the cell membrane. Moreover, the number of IL2R components in different cell lines was also estimated. It was obtained a variable distribution in the number of IL2R components depending on the cell type and the activation state. Of most significance, the study predicts that not only the number of IL2Rα and IL2Rß, but also the number of γc determine the capacity of the cell to capture and retain IL2 in signalling complexes. Moreover, it is also showed that different cells might use different pathways to bind IL2 as consequence of its IL2R components distribution in the membrane.


Subject(s)
Interleukin Receptor Common gamma Subunit/metabolism , Interleukin-2/metabolism , Algorithms , Calibration , Cell Line , Cell Membrane/metabolism , Humans , Kinetics , Ligands , Models, Statistical , Models, Theoretical , Protein Binding , Protein Multimerization , Signal Transduction
7.
Front Immunol ; 4: 439, 2013 Dec 11.
Article in English | MEDLINE | ID: mdl-24376444

ABSTRACT

Several reports in the literature have drawn a complex picture of the effect of treatments aiming to modulate IL2 activity in vivo. They seem to promote either immunity or tolerance, probably depending on the specific context, dose, and timing of their application. Such complexity might derive from the pleiotropic role of IL2 in T cell dynamics. To theoretically address the latter possibility, our group has developed several mathematical models for Helper, Regulatory, and Memory T cell population dynamics, which account for most well-known facts concerning their relationship with IL2. We have simulated the effect of several types of therapies, including the injection of: IL2; antibodies anti-IL2; IL2/anti-IL2 immune-complexes; and mutant variants of IL2. We studied the qualitative and quantitative conditions of dose and timing for these treatments which allow them to potentiate either immunity or tolerance. Our results provide reasonable explanations for the existent pre-clinical and clinical data, predict some novel treatments, and further provide interesting practical guidelines to optimize the future application of these types of treatments.

8.
J Immunol ; 190(12): 6230-8, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23677467

ABSTRACT

IL-2 has been used for the treatment of melanoma and renal cell carcinoma, but this therapy has limited efficacy and severe toxicity. Currently, it is assumed that part of the limited efficacy is due to the IL-2-driven preferential expansion of regulatory T cells, which dampen the antitumor immunity. In this study, we characterize a human IL-2 mutant with higher antitumor efficacy and lower toxicity than wild type human IL-2 (wtIL-2). The mutant differs from wtIL-2 by four mutations at the interface with the α subunit of IL-2R. The IL-2 mutant induces in vitro proliferation of CD8(+)CD44(hi) and NK1.1 cells as efficiently as does wtIL-2, but it shows a reduced capacity to induce proliferation of CD4(+)Foxp3(+) regulatory T cells. The IL-2 mutant shows a higher antimetastatic effect than does wtIL-2 in several transplantable tumor models: the experimental metastasis model of MB16F0 melanoma and the experimental and spontaneous metastasis models for the mouse pulmonary carcinoma 3LL-D1222. Relevantly, the IL-2 mutant also exhibits lower lung and liver toxicity than does wtIL-2 when used at high doses in mice. In silico simulations, using a calibrated mathematical model, predict that the properties of IL-2 mutein are a consequence of the reduction, of at least two orders of magnitude, in its affinity for the α subunit of IL-2R (CD25). The human IL-2 mutant described in the present work could be a good candidate for improving cancer therapy based on IL-2.


Subject(s)
Immunotherapy/methods , Interleukin-2/genetics , Interleukin-2/immunology , Neoplasms, Experimental/therapy , Animals , Disease Models, Animal , Flow Cytometry , Humans , Interleukin-2/chemistry , Melanoma/immunology , Melanoma/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Theoretical , Mutation , Neoplasms, Experimental/immunology , Protein Structure, Quaternary
9.
Int Immunol ; 24(7): 427-46, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22371423

ABSTRACT

Several reports in the literature have drawn a complex picture of the effect of treatments aiming to modulate IL2 activity in vivo. They seem to promote indistinctly immunity or tolerance, probably depending on the specific context, dose and timing of their application. Such complexity might derives from the dual role of IL2 on T-cell dynamics. To theoretically address the latter possibility, we develop a mathematical model for helper, regulatory and memory T-cells dynamics, which account for most well-known facts relative to their relationship with IL2. We simulate the effect of three types of therapies: IL2 injections, IL2 depletion using anti-IL2 antibodies and IL2/anti-IL2 immune complexes injection. We focus in the qualitative and quantitative conditions of dose and timing for these treatments which allow them to potentate either immunity or tolerance. Our results provide reasonable explanations for the existent pre-clinical and clinical data and further provide interesting practical guidelines to optimize the future application of these types of treatments. Particularly, our results predict that: (i) Immune complexes IL2/anti-IL2 mAbs, using mAbs which block the interaction of IL2 and CD25 (the alpha chain of IL2 receptor), is the best option to potentate immunity alone or in combination with vaccines. These complexes are optimal when a 1:2 molar ratio of mAb:IL2 is used and the mAbs have the largest possible affinity; (ii) Immune complexes IL2/anti-IL2 mAbs, using mAbs which block the interaction of IL2 and CD122 (the beta chain of IL2 receptor), are the best option to reinforce preexistent natural tolerance, for instance to prevent allograft rejection. These complexes are optimal when a 1:2 molar ratio of mAb:IL2 is used and the mAbs have intermediate affinities; (iii) mAbs anti-IL2 can be successfully used alone to treat an ongoing autoimmune disorder, promoting the re-induction of tolerance. The best strategy in this therapy is to start treatment with an initially high dose of the mAbs (one capable to induce some immune suppression) and then scales down slowly the dose of mAb in subsequent applications.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immunotherapy , Models, Immunological , T-Lymphocytes, Regulatory/immunology , Animals , Cell Communication , Computer Simulation , Humans , Immune Tolerance , Immunologic Memory , Interleukin-2/antagonists & inhibitors , Interleukin-2/immunology , Interleukin-2/therapeutic use , Lymphocyte Activation
10.
J Theor Biol ; 262(4): 720-32, 2010 Feb 21.
Article in English | MEDLINE | ID: mdl-19878686

ABSTRACT

Mathematical models accounting for well-known evidences relating to the dynamics of interleukin 2, helper and regulatory T cells are presented. These models extend an existent model (the so-called cross-regulation model of immunity), by assuming IL-2 as the growth factor produced by helper cells, but used by both helper and regulatory cells to proliferate and survive. Two model variants, motivated by current literature, are explored. The first variant assumes that regulatory cells suppress helper cells by limiting IL-2 production and consuming the available IL-2; i.e. they just trigger competition for IL-2. The second model variant adds to the latter competitive mechanism the direct inhibition of helper cells activation by regulatory cells. The extended models retain key dynamical features of the cross-regulation model. But such reasonable behavior depends on parameter constraints, which happen to be realistic and lead to interesting biological discussions. Furthermore, the introduction of IL-2 in these models breaks the local/specific character of interactions, providing new properties to them. In the extended models, but not in the cross-regulation model, the response triggered by an antigen affects the response to other antigens in the same lymph node. The first model variant predicts an unrealistic coupling of the immune reactions to all the antigens in the lymph node. In contrast, the second model variant allows the coexistent of concomitant tolerant and immune responses to different antigens. The IL-2 derived from an ongoing immune reaction reinforces tolerance to other antigens in the same lymph node. Overall the models introduced here are useful extensions of the cross-regulation formalism. In particular, they might allow future studies of the effect of different IL-2 modulation therapies on CD4+ T cell dynamics.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Interleukin-2/physiology , T-Lymphocytes, Helper-Inducer/metabolism , Animals , Antigen Presentation , CD4-Positive T-Lymphocytes/cytology , Cell Proliferation , Computer Simulation , Humans , Immune System , Interleukin-2/metabolism , Kinetics , Lymph Nodes/pathology , Mice , Mice, Knockout , Models, Biological , Signal Transduction , T-Lymphocytes, Helper-Inducer/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...