Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters

Publication year range
1.
Int Immunol ; 33(12): 749-754, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34174067

ABSTRACT

Semaphorins were originally identified as axon-guidance molecules essential for neural development. In addition to their functions in the neural system, members of the semaphorin family have critical functions in many pathophysiological processes, including immune responses, bone homeostasis, cancer and metabolic disorders. In particular, several lines of evidence indicate that mammalian/mechanistic target of rapamycin (mTOR), a central regulator of cell metabolism, regulates the functions of semaphorins in various types of cells, revealing a novel link between semaphorins and cell metabolism. In this review, we discuss recent advances in the immunometabolic functions of semaphorins, with a particular focus on mTOR signaling.


Subject(s)
Nerve Tissue Proteins , Semaphorins , Animals , Humans , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/metabolism , Semaphorins/immunology , Semaphorins/metabolism , Signal Transduction/immunology , Sirolimus/immunology , Sirolimus/metabolism
2.
Int J Cancer ; 143(11): 3008-3018, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30183073

ABSTRACT

mTOR pathway inhibitors such as rapalogs represent a promising tool to induce functional memory CD8 T cells. In our study, we investigated the combination of temsirolimus with anticancer vaccines. Using various designs of cancer vaccines (short and long peptides or the B subunit of Shiga toxin as an antigen delivery vector) and tumor models (melanoma, lung and colon cancer), we showed that the administration of temsirolimus efficiently decreased tumor growth and enhanced tumor-specific CD8 T-cell responses induced by vaccination. Furthermore, tumor-specific CD8 T cells induced by the bi-therapy (vaccine + temsirolimus) exhibit phenotypic characteristics of central memory (CD127+ CD62L+ ) CD8 T cells compared to vaccination alone. We demonstrated that regulatory CD4 T cells (Tregs ) expansion in vivo limits the efficacy of the bi-therapy by altering the antitumor CD8 T-cell responses. Finally, the use of a small molecule CCR4 antagonist to prevent Tregs induction considerably improved the efficacy of the bi-therapy by enhancing CD8 T cells-mediated antitumor immunity. Taken together, our study highlights the potential interest of combining cancer vaccines with drugs that promote memory CD8 T cells and inhibit Tregs .


Subject(s)
Cancer Vaccines/immunology , Receptors, CCR4/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Immunologic Memory/drug effects , Immunologic Memory/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Sirolimus/analogs & derivatives , Sirolimus/immunology , Sirolimus/pharmacology , Vaccination/methods
3.
Immunity ; 30(6): 759-61, 2009 Jun 19.
Article in English | MEDLINE | ID: mdl-19538925

ABSTRACT

The decision of naive Th cells to assume the fate of effector or regulator Th cells heavily influences the outcome of adaptive immune responses. In this issue of Immunity, Delgoffe et al. (2009) use genetic approaches and demonstrate that mTOR kinase plays a critical role in this decision-making process.


Subject(s)
Carrier Proteins/metabolism , Cell Differentiation/immunology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology , Transcription Factors/immunology , Animals , Carrier Proteins/genetics , Cytokines/immunology , Cytokines/metabolism , Mice , Phosphotransferases (Alcohol Group Acceptor)/genetics , Sirolimus/immunology , Sirolimus/metabolism , T-Lymphocyte Subsets/enzymology , T-Lymphocytes, Helper-Inducer/enzymology , TOR Serine-Threonine Kinases , Transcription Factors/metabolism
4.
J Immunol ; 195(5): 2038-45, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26238486

ABSTRACT

CD1d-restricted activation of invariant NKT (iNKT) cells results in the abundant production of various types of cytokines and the subsequent modulation of immune responses. This has been shown to be relevant in several clinical disorders, including cancer, autoimmunity, and graft tolerance. Although it is well known that the suppressive function of regulatory T cells is critically dependent on the FOXP3 gene, FOXP3 can also be expressed by conventional human T cells upon activation, indicating the lack of specificity of FOXP3 as a marker for suppressive cells. In this study, we report that the mammalian target of rapamycin (mTOR) inhibitor rapamycin and IL-10, but not TGF-ß, can induce FOXP3 expression in iNKT cell lines. Importantly, however, FOXP3(+) iNKT cells only acquired suppressive abilities when cultured in the presence of the mTOR inhibitor rapamycin. Suppression of responder T cell proliferation by FOXP3(+) iNKT cells was found to be cell contact-dependent and was accompanied by a reduced capacity of iNKT cells to secrete IFN-γ. Notably, imaging flow cytometry analysis demonstrated predominant nuclear localization of FOXP3 in suppressive FOXP3(+) iNKT cells, whereas nonsuppressive FOXP3(+) iNKT cells showed a predominance of cytoplasmically localized FOXP3. In conclusion, whereas IL-10 can enhance FOXP3 expression in iNKT cells, mTOR inhibition is solely required for promoting nuclear localization of FOXP3 and the induction of suppressive FOXP3(+) iNKT cells.


Subject(s)
Cell Nucleus/immunology , Forkhead Transcription Factors/immunology , Natural Killer T-Cells/immunology , TOR Serine-Threonine Kinases/immunology , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/immunology , CTLA-4 Antigen/immunology , CTLA-4 Antigen/metabolism , Cell Line , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Flow Cytometry , Forkhead Transcription Factors/metabolism , Humans , Immunosuppressive Agents/immunology , Immunosuppressive Agents/pharmacology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-10/immunology , Interleukin-10/pharmacology , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Natural Killer T-Cells/metabolism , Sirolimus/immunology , Sirolimus/pharmacology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism
5.
Am J Respir Crit Care Med ; 193(2): 143-53, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26426522

ABSTRACT

RATIONALE: Corticosteroid resistance is a major barrier to the effective treatment of chronic obstructive pulmonary disease (COPD). Several molecular mechanisms have been proposed, such as activations of the phosphoinositide-3-kinase/Akt pathway and p38 mitogen-activated protein kinase. However, the mechanism for corticosteroid resistance is still not fully elucidated. OBJECTIVES: To investigate the role of mammalian target of rapamycin (mTOR) in corticosteroid sensitivity in COPD. METHODS: The corticosteroid sensitivity of peripheral blood mononuclear cells collected from patients with COPD, smokers, and nonsmoking control subjects, or of human monocytic U937 cells exposed to cigarette smoke extract (CSE), was quantified as the dexamethasone concentration required to achieve 30% inhibition of tumor necrosis factor-α-induced CXCL8 production in the presence or absence of the mTOR inhibitor rapamycin. mTOR activity was determined as the phosphorylation of p70 S6 kinase, using Western blotting. MEASUREMENTS AND MAIN RESULTS: mTOR activity was increased in peripheral blood mononuclear cells from patients with COPD, and treatment with rapamycin inhibited this as well as restoring corticosteroid sensitivity. In U937 cells, CSE stimulated mTOR activity and c-Jun expression, but pretreatment with rapamycin inhibited both and also reversed CSE-induced corticosteroid insensitivity. CONCLUSIONS: mTOR inhibition by rapamycin restores corticosteroid sensitivity via inhibition of c-Jun expression, and thus mTOR is a potential novel therapeutic target for COPD.


Subject(s)
Adrenal Cortex Hormones/pharmacology , Drug Resistance/drug effects , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-jun/drug effects , Pulmonary Disease, Chronic Obstructive/drug therapy , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/drug effects , Adrenal Cortex Hormones/therapeutic use , Aged , Drug Resistance/immunology , Female , Histone Deacetylase 2/drug effects , Histone Deacetylase 2/physiology , Humans , Immunosuppressive Agents/immunology , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Male , Middle Aged , Oxidative Stress/physiology , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/physiology , Proto-Oncogene Proteins c-jun/physiology , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/physiopathology , Sirolimus/immunology , Sirolimus/therapeutic use , Smoking/adverse effects , Smoking/physiopathology , TOR Serine-Threonine Kinases/physiology , U937 Cells/drug effects , p38 Mitogen-Activated Protein Kinases/drug effects , p38 Mitogen-Activated Protein Kinases/physiology
6.
Cell Immunol ; 301: 74-81, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26687613

ABSTRACT

The immune response of hemophilia A patients to administered FVIII is a major complication that obviates this very therapy. We have recently described the use of synthetic, biodegradable nanoparticles carrying rapamycin and FVIII peptide antigens, to induce antigen-specific tolerance. Herein we test the tolerogenicity of nanoparticles that contains full length FVIII protein in hemophilia A mice, focusing on anti-FVIII humoral immune response. As expected, recipients of tolerogenic nanoparticles remained unresponsive to FVIII despite multiple challenges for up to 6 months. Furthermore, therapeutic treatments in FVIII-immunized mice with pre-existing anti-FVIII antibodies resulted in diminished antibody titers, albeit efficacy required longer therapy with the tolerogenic nanoparticles. Interestingly, durable FVIII-specific tolerance was also achieved in animals co-administered with FVIII admixed with nanoparticles encapsulating rapamycin alone. These results suggest that nanoparticles carrying rapamycin and FVIII can be employed to induce specific tolerance to prevent and even reverse inhibitor formation.


Subject(s)
Factor VIII/administration & dosage , Hemophilia A/immunology , Immune Tolerance/immunology , Immunosuppressive Agents/administration & dosage , Nanoparticles , Sirolimus/administration & dosage , Animals , Antibodies, Neutralizing/immunology , Disease Models, Animal , Factor VIII/immunology , Immunosuppressive Agents/immunology , Mice , Sirolimus/immunology , Vaccines, Synthetic
7.
Cytotherapy ; 17(9): 1188-99, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26276002

ABSTRACT

BACKGROUND AIMS: Multipotent mesenchymal stromal cells (MSCs) are distinguished by their ability to differentiate into a number of stromal derivatives of interest for regenerative medicine, but they also have immunoregulatory properties that are being tested in a number of clinical settings. METHODS: We show that brief incubations with rapamycin, everolimus, FK506 or cyclosporine A increase the immunosuppressive potency of MSCs and other cell types. RESULTS: The treated MSCs are up to 5-fold more potent at inhibiting the induced proliferation of T lymphocytes in vitro. We show that this effect probably is due to adsorption of the drug by the MSCs during pre-treatment, with subsequent diffusion into co-cultures at concentrations sufficient to inhibit T-cell proliferation. MSCs contain measurable amounts of rapamycin after a 15-min exposure, and the potentiating effect is blocked by a neutralizing antibody to the drug. With the use of a pre-clinical model of acute graft-versus-host disease, we demonstrate that a low dose of rapamycin-treated but not untreated umbilical cord-derived MSCs significantly inhibit the onset of disease. CONCLUSIONS: The use of treated MSCs may achieve clinical end points not reached with untreated MSCs and allow for infusion of fewer cells to reduce costs and minimize potential side effects.


Subject(s)
Graft vs Host Disease/prevention & control , Immune Tolerance/drug effects , Immunosuppressive Agents/pharmacology , Mesenchymal Stem Cell Transplantation/methods , Sirolimus/pharmacology , Animals , Antibodies, Neutralizing/immunology , Cell Proliferation/drug effects , Coculture Techniques , Cyclosporine/pharmacology , Disease Models, Animal , Everolimus/pharmacology , Female , Graft vs Host Disease/immunology , Humans , Immunosuppression Therapy/methods , Lymphocyte Activation/immunology , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/immunology , Mice , Mice, Inbred BALB C , Sirolimus/immunology , T-Lymphocytes/immunology , Tacrolimus/pharmacology , Umbilical Cord/cytology
8.
Genes Immun ; 15(6): 413-23, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24898389

ABSTRACT

The induction of functional memory cytotoxic T lymphocytes (CTLs) is a major goal of vaccination against intracellular pathogens. Interleukin (IL)-12 is critical for the generation of memory CTLs, and inhibition of mammalian target of rapamycin (mTOR) by rapamycin can effectively enhance the memory CTL response. Yet, the role of IL-12 in mTOR's regulation of memory CTL is unknown. Here we hypothesized that the immunostimulatory effects of mTOR on memory CTLs requires IL-12 signaling. Our results revealed that rapamycin increased the generation of memory CTLs in vaccinia virus infection, and this enhancement was dependent upon the IL-12 signal. Furthermore, IL-12 receptor deficiency diminished the secondary expansion of rapamycin-regulated memory and resultant secondary memory CTLs were abolished. Rapamycin enhanced IL-12 signaling by upregulating IL-12 receptor ß2 expression and signal transducer and activator of transcription factor 4 phosphorylation in CTLs during early infection. In addition, rapamycin continually suppressed T-bet expression in both wild-type and IL-12 receptor knockout CTLs. These results indicate an essential role for IL-12 in the regulation of memory CTLs by mTOR and highlight the importance of considering the interplay between cytokines and adjuvants during vaccine design.


Subject(s)
Immunologic Memory/immunology , Interleukin-2/immunology , T-Lymphocytes, Cytotoxic/immunology , TOR Serine-Threonine Kinases/immunology , Vaccinia virus/immunology , Vaccinia/immunology , Adoptive Transfer , Animals , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Flow Cytometry , Host-Pathogen Interactions/immunology , Immunosuppressive Agents/immunology , Immunosuppressive Agents/pharmacology , Interleukin-2/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Interleukin-12/deficiency , Receptors, Interleukin-12/genetics , Receptors, Interleukin-12/immunology , STAT4 Transcription Factor/immunology , STAT4 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Sirolimus/immunology , Sirolimus/pharmacology , T-Box Domain Proteins/immunology , T-Box Domain Proteins/metabolism , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/metabolism , TOR Serine-Threonine Kinases/metabolism , Vaccinia/genetics , Vaccinia/virology , Vaccinia virus/physiology
9.
Biol Blood Marrow Transplant ; 20(2): 192-201, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24239650

ABSTRACT

Rapamycin (RAPA) inhibits the mechanistic target of rapamycin (mTOR), a crucial immune system regulator. Dendritic cells (DC) generated in RAPA (RAPA-DC) enrich for CD4(+) forkhead box p3 (FoxP3(+)) regulatory T cells and induce T cell apoptosis by an unknown mechanism. RAPA-DC also promote experimental allograft survival, yet paradoxically secrete increased IL-12, crucial for the generation of IFN-γ(+) CD4(+) T cells. However, IFN-γ is pro-apoptotic and IL-12-driven IFN-γ inhibits experimental graft-versus-host disease (GVHD). We hypothesized that IL-12(hi) RAPA-DC would facilitate IFN-γ-mediated apoptosis of alloreactive T cells and, unlike control (CTR)-DC, would reduce lethal GVHD. Following LPS stimulation, RAPA-DC exhibited decreased MHCII and co-stimulatory molecules and contained a significant population of CD86(lo) IL-12(hi) cells. Consistent with our hypothesis, both unstimulated and LPS-stimulated RAPA-DC enhanced alloreactive CD4(+) T cell apoptosis in culture. Augmented T cell apoptosis was ablated by IFN-γ neutralization or using T cells lacking the IFN-γ receptor, and it was associated with increased expression of Fas and cleaved caspase 8. DC production or responses to IFN-γ were not important to increased apoptotic functions of RAPA-DC. LPS-stimulated IL-12p40(-/-) RAPA-DC induced lower levels of T cell apoptosis in culture, which was further decreased with addition of anti-IFN-γ. Finally, whereas CTR-DC accelerated mortality from GVHD, LPS-treated RAPA-DC significantly prolonged host survival. In conclusion, increased apoptosis of allogeneic CD4(+) T cells induced by LPS-stimulated IL-12(hi) RAPA-DC is mediated in vitro through IFN-γ and in part by increased IL-12 expression. Enhanced production of IL-12, the predominant inducer of IFN-γ by immune cells, is a probable mechanism underlying the capacity of LPS-treated RAPA-DC to reduce GVHD.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Apoptosis/immunology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Graft vs Host Disease/immunology , Interleukin-12/immunology , Sirolimus/therapeutic use , Animals , Antibiotics, Antineoplastic/immunology , Cell Differentiation , Cell Proliferation , Female , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Sirolimus/immunology
10.
Clin Immunol ; 153(2): 332-42, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24909732

ABSTRACT

The golden standard for functional evaluation of immunodeficiencies is the incorporation of [(3)H]-thymidine in a proliferation assay stimulated with mitogens. Recently developed whole blood proliferation assays have the advantage of parallel lymphocyte lineage analysis and in addition provide a non-radioactive alternative. Here we evaluate the Flow-cytometric Assay for Specific Cell-mediated Immune-response in Activated whole blood (FASCIA) in a comparison with [(3)H]-thymidine incorporation in four patients with severe combined immunodeficiency. The threshold for the minimum number of lymphocytes required for reliable responses in FASCIA is determined together with reference values from 100 healthy donors when stimulated with mitogens as well as antigen specific stimuli. Finally, responses against PWM and SEA+SEB stimuli are conducted with clinically relevant immunomodulatory compounds. We conclude that FASCIA is a rapid, stable and sensitive functional whole blood assay that requires small amounts of whole blood that can be used for reliable assessment of lymphocyte reactivity in patients.


Subject(s)
B-Lymphocytes/immunology , Cell Proliferation , Flow Cytometry/methods , Severe Combined Immunodeficiency/immunology , T-Lymphocytes/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/pathology , Dexamethasone/immunology , Dexamethasone/pharmacology , Enterotoxins/immunology , Enterotoxins/pharmacology , Humans , Immunosuppressive Agents/immunology , Immunosuppressive Agents/pharmacology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-17/immunology , Interleukin-17/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocyte Count , Pokeweed Mitogens/immunology , Pokeweed Mitogens/pharmacology , Reproducibility of Results , Severe Combined Immunodeficiency/diagnosis , Sirolimus/immunology , Sirolimus/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/pathology , Tacrolimus/immunology , Tacrolimus/pharmacology
11.
Liver Int ; 33(5): 656-65, 2013 May.
Article in English | MEDLINE | ID: mdl-23442173

ABSTRACT

Current strategies for immunosuppression in liver transplant (LT) recipients include the design of protocols targeting a more individualized approach to reduce risk factors such as renal failure, cardiovascular complications and malignancies. Renal injury in LT recipients may be often multifactorial and is associated with increased risk of post-transplant morbidity and mortality. The quest for low toxicity immunosuppressive regimens has been challenging and resulted in CNI minimization protocols or CNI withdrawal and conversion to mycophenolate mofetil (MMF) and/or mammalian target of rapamycin inhibitor-based immunosuppressive regimens. Use of antibody induction to delay CNI administration may be an option in particular in immunocompromized, critically ill patients with high MELD scores. Protocols including MMF introduction and concomitant CNI minimization have the potential to recover renal function even in the medium and long term after LT. We review on hot topics in the prevention and management of acute and chronic renal injury in LT patients. For this purpose, we present and critically discuss results from immunosuppressive studies published in the current literature or presented at recent LT meetings.


Subject(s)
Calcineurin Inhibitors , Immunosuppression Therapy/methods , Liver Transplantation/immunology , Precision Medicine/methods , Renal Insufficiency/prevention & control , Abatacept , Everolimus , Humans , Immunoconjugates/immunology , Immunoconjugates/pharmacology , Liver Transplantation/adverse effects , Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/immunology , Mycophenolic Acid/pharmacology , Pyrroles/immunology , Pyrroles/pharmacology , Quinazolines/immunology , Quinazolines/pharmacology , Renal Insufficiency/etiology , Risk Factors , Sirolimus/analogs & derivatives , Sirolimus/immunology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/immunology , Tacrolimus/immunology , Tacrolimus/pharmacology
13.
Blood ; 116(24): 5403-18, 2010 Dec 09.
Article in English | MEDLINE | ID: mdl-20833977

ABSTRACT

We have developed a major histocompatibility complex-defined primate model of graft-versus-host disease (GVHD) and have determined the effect that CD28/CD40-directed costimulation blockade and sirolimus have on this disease. Severe GVHD developed after haploidentical transplantation without prophylaxis, characterized by rapid clinical decline and widespread T-cell infiltration and organ damage. Mechanistic analysis showed activation and possible counter-regulation, with rapid T-cell expansion and accumulation of CD8(+) and CD4(+) granzyme B(+) effector cells and FoxP3(pos)/CD27(high)/CD25(pos)/CD127(low) CD4(+) T cells. CD8(+) cells down-regulated CD127 and BCl-2 and up-regulated Ki-67, consistent with a highly activated, proliferative profile. A cytokine storm also occurred, with GVHD-specific secretion of interleukin-1 receptor antagonist (IL-1Ra), IL-18, and CCL4. Costimulation Blockade and Sirolimus (CoBS) resulted in striking protection against GVHD. At the 30-day primary endpoint, CoBS-treated recipients showed 100% survival compared with no survival in untreated recipients. CoBS treatment resulted in survival, increasing from 11.6 to 62 days (P < .01) with blunting of T-cell expansion and activation. Some CoBS-treated animals did eventually develop GVHD, with both clinical and histopathologic evidence of smoldering disease. The reservoir of CoBS-resistant breakthrough immune activation included secretion of interferon-γ, IL-2, monocyte chemotactic protein-1, and IL-12/IL-23 and proliferation of cytotoxic T-lymphocyte-associated antigen 4 immunoglobulin-resistant CD28(-) CD8(+) T cells, suggesting adjuvant treatments targeting this subpopulation will be needed for full disease control.


Subject(s)
CD28 Antigens , CD8-Positive T-Lymphocytes/immunology , Graft vs Host Disease/prevention & control , Immunosuppression Therapy/methods , Sirolimus/therapeutic use , Animals , Cell Proliferation , Graft vs Host Disease/etiology , Graft vs Host Disease/pathology , Haplotypes , Hematopoietic Stem Cell Transplantation/adverse effects , Lymphocyte Activation , Macaca mulatta , Sirolimus/immunology
14.
J Immunol ; 185(8): 4750-9, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20844194

ABSTRACT

The critical roles of TGF-ß in the reciprocal differentiation of tolerance-promoting CD4(+)Foxp3(+) regulatory T cells (Tregs) and proinflammatory Th17 effector cells affect alloimmune reactivity and transplant outcome. We reasoned that a strategy to harness TGF-ß and block proinflammatory cytokines would inhibit the differentiation of Th17 cells and strengthen the cadre of Tregs to promote tolerance induction and long-term allograft survival. In this study, we report the development of a long-lasting autoactive human mutant TGF-ß1/Fc fusion protein that acts in conjunction with rapamycin to inhibit T cell proliferation and induce the de novo generation of Foxp3(+) Treg in the periphery, while at the same time inhibiting IL-6-mediated Th17 cell differentiation. Short-term combined treatment with TGF-ß1/Fc and rapamycin achieved long-term pancreatic islet allograft survival and donor-specific tolerance in a mouse model. This effect was accompanied by expansion of Foxp3(+) Tregs, enhanced alloantigen-specific Treg function, and modulation of transcript levels of Foxp3, IL-6, and IL-17. Our strategy of combined TGF-ß1/Fc and rapamycin to target the IL-6-related Tregs and Th17 signaling pathways provides a promising approach for inducing transplant tolerance and its clinical application.


Subject(s)
Immunosuppressive Agents/therapeutic use , Islets of Langerhans Transplantation/immunology , Recombinant Fusion Proteins/therapeutic use , Sirolimus/therapeutic use , Transforming Growth Factor beta1/immunology , Transplantation Tolerance/immunology , Animals , Blotting, Western , Cell Differentiation/immunology , Cell Separation , Flow Cytometry , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/immunology , Gene Knock-In Techniques , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , Immunohistochemistry , Immunosuppressive Agents/immunology , Interleukin-17/biosynthesis , Interleukin-17/immunology , Interleukin-6/biosynthesis , Interleukin-6/immunology , Male , Mice , Mice, Inbred C57BL , Mutation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Sirolimus/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta1/genetics , Transplantation, Homologous
15.
Am J Transplant ; 11(3): 613-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21342450

ABSTRACT

Sirolimus is a potent antiproliferative agent used clinically to prevent renal allograft rejection. However, little is known about the effects of maintenance immunosuppressive agents on the immune response to potentially protective vaccines. Here we show that sirolimus paradoxically increases the magnitude and quality of the CD8+ T-cell response to vaccinia vaccination in nonhuman primates, fostering more robust recall responses compared to untreated and tacrolimus-treated controls. Enhancement of both the central and effector memory compartments of the vaccinia-specific CD8+ T-cell response was observed. These data elucidate new mechanistic characteristics of sirolimus and suggest immune applications extending beyond its role as an immunosuppressant.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Sirolimus/therapeutic use , Vaccinia/prevention & control , Viral Vaccines/therapeutic use , Animals , CD8-Positive T-Lymphocytes/virology , Cytokines/metabolism , Flow Cytometry , Immunologic Memory/drug effects , Immunosuppressive Agents/immunology , Immunosuppressive Agents/therapeutic use , Macaca mulatta , Sirolimus/immunology , Vaccination , Vaccinia/immunology , Vaccinia/virology , Vaccinia virus/immunology , Viral Vaccines/immunology
16.
Clin Lab ; 57(5-6): 403-6, 2011.
Article in English | MEDLINE | ID: mdl-21755832

ABSTRACT

BACKGROUND: The aim of our study was to evaluate the possible determination of everolimus concentrations using the newly-introduced sirolimus antibody conjugated magnetic immunoassay (ACMIA). METHODS: Everolimus concentrations were determined in 100 blood samples from kidney (n = 47) and liver (n = 53) transplant recipients using the IMx sirolimus microparticle enzyme immunoassay (MEIA) from Abbott as previously described (Clin Biochem 2007;40:132-36) and sirolimus ACMIA from Siemens Healthcare Diagnostics Ltd. RESULTS: The ACMIA everolimus values were significantly higher than those of MEIA (p < 0.001). Analogous slope and intercept values were obtained in the linear regression between the ACMIA and MEIA results when compared to the Seradyn Certican everolimus controls or the blood samples from transplant recipients. Correction of the ACMIA values using the regression equation obtained for the control material (ACMIAcorrected = 0.55 ACMIA + 1.14) led to a satisfactory relationship with the results provided by the MEIA for the patients' samples (MEIA = 1.00 ACMIAcorrected + 0.30, r = 0.905, p < 0.001). CONCLUSIONS: The sirolimus ACMIA on the Dimension platform, which does not require manual pre-treatment of the blood samples, may be an acceptable option for therapeutic everolimus monitoring, significantly reducing technician time in comparison to other widely-used immunoassays.


Subject(s)
Drug Monitoring/methods , Immunomagnetic Separation/methods , Immunosuppressive Agents/blood , Kidney Transplantation , Liver Transplantation , Sirolimus/analogs & derivatives , Sirolimus/immunology , Antibody Specificity , Cross Reactions , Everolimus , Humans , Immunoenzyme Techniques , Reproducibility of Results , Sensitivity and Specificity , Sirolimus/blood , Time Factors
17.
Anal Biochem ; 407(2): 160-4, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-20696124

ABSTRACT

Dot-blot is a versatile and simple analysis to perform. We adapted this method as a simple identity test for monoclonal antibodies to a number of small compounds: three transplant drugs, an anticonvulsant, a steroid, an anticancer drug, and an antibiotic. Immunology-based identity tests using low-molecular-mass organic compounds have historically been a challenge to develop. We modified the traditional dot-blot assay to serve as an identity test for monoclonal antibodies to carbamazepine, sirolimus, tacrolimus, cyclosporine, cortisol, methotrexate, and gentamicin. The primary obstacle was the immobilization of these organic compounds on nitrocellulose as nitrocellulose is also soluble in most of the organic solvents in which the compounds are soluble. We evaluated different membranes, solvents, and chemical forms of these organic compounds to overcome this challenge. A number of incubation and washing solutions were also investigated. By varying the chemical form, concentration, and incubation conditions, a set of effective and reproducible identity tests were developed for these monoclonal antibodies.


Subject(s)
Antibodies, Monoclonal , Immunoblotting/methods , Pharmaceutical Preparations/analysis , Acridines/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/immunology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/immunology , Antibodies, Monoclonal/chemistry , Anticonvulsants/chemistry , Anticonvulsants/immunology , Carbamazepine/chemistry , Carbamazepine/immunology , Collodion/chemistry , Cyclosporine/chemistry , Cyclosporine/immunology , Gentamicins/chemistry , Gentamicins/immunology , Hydrocortisone/chemistry , Hydrocortisone/immunology , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/immunology , Methotrexate/chemistry , Methotrexate/immunology , Reproducibility of Results , Sirolimus/chemistry , Sirolimus/immunology , Tacrolimus/chemistry , Tacrolimus/immunology
18.
Mol Pharm ; 7(6): 2297-309, 2010 Dec 06.
Article in English | MEDLINE | ID: mdl-20958081

ABSTRACT

Lymphocytes are central to the progression of autoimmune disease, transplant rejection, leukemia, lymphoma and lymphocyte-resident viral diseases such as HIV/AIDs. Strategies to target drug treatments to lymphocytes, therefore, represent an opportunity to enhance therapeutic outcomes in disease states where many current treatment regimes are incompletely effective and promote significant toxicities. Here we demonstrate that highly lipophilic drug candidates that preferentially access the intestinal lymphatics after oral administration show significantly enhanced access to lymphocytes leading to improved immunomodulatory activity. When coadministered with such drugs, lipids enhance lymphocyte targeting via a three tiered action: promotion of drug absorption from the gastrointestinal tract, enhancement of lymphatic drug transport and stimulation of lymphocyte recruitment into the lymphatics. This strategy has been exemplified using a highly lipophilic immunosuppressant (JWH015) where coadministration with selected lipids led to significant increases in lymphatic transport, lymphocyte targeting and IL-4 and IL-10 expression in CD4+ and CD8+ lymphocytes after ex vivo mitogen stimulation. In contrast, administration of a 2.5-fold higher dose of JWH015 in a formulation that did not stimulate lymph transport had no effect on antiinflammatory cytokine levels, in spite of equivalent drug exposure in the blood. The current data suggest that complementary drug design and delivery strategies that combine highly lipophilic, lymphotropic drug candidates with lymph-directing formulations provide enhanced selectivity, potency and therapeutic potential for drug candidates with lymphocyte associated targets.


Subject(s)
Immunomodulation , Indoles/pharmacology , Lymphocytes/drug effects , Animals , Chromatography, High Pressure Liquid , Cytokines/biosynthesis , DDT/analysis , DDT/immunology , DDT/pharmacology , Drug Delivery Systems , Flow Cytometry , Indoles/analysis , Indoles/immunology , Lymphocytes/immunology , Male , Mitogens/chemistry , Mitogens/immunology , Mitogens/pharmacology , Phenanthrenes/analysis , Phenanthrenes/immunology , Phenanthrenes/pharmacology , Rats , Rats, Sprague-Dawley , Sirolimus/analysis , Sirolimus/immunology , Sirolimus/pharmacology
19.
Poult Sci ; 99(1): 416-422, 2020 Jan.
Article in English | MEDLINE | ID: mdl-32416826

ABSTRACT

This study was conducted to explore the regulatory role of the target of rapamycin complex 1 (TORC1) signaling pathway in crop milk synthesis in breeding pigeons (Columba livia). Three groups of breeding pigeons in the lactation period (n = 30 pairs/group) were respectively injected with rapamycin (RAPA, a specific inhibitor of the target of rapamycin complex) at doses of 0 (vehicle, control), 0.6, or 1.2 mg/kg body weight (BW)/day via the wing vein for 7 days. The average daily feed intake (ADFI) and BW of the breeding pigeons and the BW of young squabs were respectively recorded throughout the experimental period. The breeding pigeons were sacrificed to collect their crop tissues, crop milk, and serum on the eighth day of the experiment. The results showed that neither 0.6 nor 1.2 mg/kg BW RAPA injection affected BW loss or ADFI in breeding pigeons (P > 0.05), while crop thickness and crop relative weight were significantly decreased (P < 0.05) in the 1.2 mg/kg BW rapamycin-injected group. Simultaneously, RAPA (especially at 1.2 mg/kg BW) decreased the crude protein, αs1-casein, αs2-casein, ß-casein, and amino acid contents (Asp, Thr, Ser, Glu, Gly, Ala, Cys, Val, Met, Ile, Leu, Tyr, Lys, His, Arg, and Pro) of crop milk (P < 0.05) and the concentrations of albumin, total protein, and uric acid in the serum of breeding pigeons (P < 0.05). Additionally, the expression of TORC1 pathway-related proteins (TORC1, S6K1, S6, 4EBP1, and eIF4E) was downregulated in the crop tissues of breeding pigeons by 0.6 or 1.2 mg/kg BW/day RAPA injection (P < 0.05). Accordingly, the average daily gain (ADG) of young squabs declined, and the mortality rate increased significantly (P < 0.05). Together, the results showed that RAPA reduced protein and amino acid levels in the crop milk of breeding pigeons and retarded young squab growth, suggesting a crucial role of TORC1 in crop milk synthesis in breeding pigeons.


Subject(s)
Avian Proteins/antagonists & inhibitors , Avian Proteins/biosynthesis , Columbidae/metabolism , Crop, Avian/metabolism , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Columbidae/growth & development , Dose-Response Relationship, Drug , Mechanistic Target of Rapamycin Complex 1/biosynthesis , Milk Proteins/biosynthesis , Random Allocation , Sirolimus/administration & dosage , Sirolimus/immunology
20.
Parasit Vectors ; 13(1): 294, 2020 Jun 09.
Article in English | MEDLINE | ID: mdl-32517744

ABSTRACT

BACKGROUND: Leishmaniases are a group of neglected tropical parasitic diseases, mainly affecting vulnerable populations of countries with poor socioeconomic status. Development of efficient vaccines is a priority due to the increasing incidence of drug resistance and toxicity to current treatments. In the search for a safe and efficient protective vaccine for human and dog visceral leishmaniases, we analyzed the suitability of the immunomodulatory drug sirolimus (SIR) to boost a preventive DNA vaccine against leishmaniasis. SIR is an already marketed drug that has been described to boost immune protection against different disease models and has also emerged as a promising therapeutic drug against L. major. METHODS: Syrian hamsters were treated with SIR concomitantly with the administration of a DNA vaccine formulation consisting in four plasmids carrying the Leishmania genes LACK, TRYP, PAPLE22 and KMPII, respectively. Two weeks after the last vaccination, the animals were infected intraperitoneally with L. infantum parasites. Five weeks post-infection the parasite load was measured by real-time PCR in target tissues and immune response was evaluated by determining anti-Leishmania specific antibodies in combination with cytokine expression in the spleen. RESULTS: Our results show that the DNA vaccine itself efficiently reduced the burden of parasites in the skin (P = 0.0004) and lymph nodes (P = 0.0452). SIR administration also enhanced the protection by reducing the parasite load in the spleen (P = 0.0004). Vaccinated animals with or without SIR co-treatment showed lower IFN-γ expression levels than those found in the spleen of control animals. mRNA expression levels of NOS2 and IL-10 were found to be significantly higher in the vaccinated plus SIR treated group. CONCLUSIONS: Co-administration of SIR enhances a DNA vaccination regimen against L. infantum, improving the reduction of parasite load in skin, lymph node and spleen. The analysis of immune markers in the spleen after challenge suggests that the trend to recover naïve levels of IFN-γ and IL-10, and the concurrent higher expression of NOS2, may be responsible for the protection induced by our vaccine co-administered with SIR.


Subject(s)
Antibodies, Protozoan/blood , Immunologic Factors/administration & dosage , Leishmaniasis, Visceral/prevention & control , Sirolimus/administration & dosage , Vaccines, DNA/immunology , Animals , Cricetinae , Cytokines/immunology , Immunization , Immunologic Factors/immunology , Leishmania infantum , Leishmaniasis, Visceral/immunology , Male , Mesocricetus , Parasite Load , Plasmids/genetics , Protozoan Proteins/immunology , Sirolimus/immunology , Spleen/immunology , Spleen/parasitology , Vaccines, DNA/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL