Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 47
1.
J Leukoc Biol ; 2024 Apr 17.
Article En | MEDLINE | ID: mdl-38630873

Regulatory T cells (Tregs) play a crucial role in the homeostasis of the immune response. Tregs are mainly generated in the thymus and are characterized by the expression of Foxp3, which is considered the Treg master transcription factor. In addition, Tregs can be induced from naïve CD4+ T cells to express Foxp3 under specific conditions both in vivo (pTregs) and in vitro (iTregs). Both subsets tTregs and pTregs are necessary for the establishment of immune tolerance to self and non-self antigens. Although it has been postulated that iTregs may be less stable compared to tTregs, mainly due to epigenetic differences, accumulating evidence in animal models shows that iTregs are stable in vivo and could be used for the treatment of inflammatory disorders including autoimmune diseases and allogeneic transplant rejection. In this review, we describe the biological characteristics of induced T regs, the key factors involved in iTreg transcriptional, metabolic and epigenetic regulation and discuss recent advances for de novo generation of stable Tregs and their use as immunotherapeutic tools in different experimental models. Moreover, we discuss the challenges and considerations for the application of iTregs in clinical trials and describe the new approaches proposed to achieve in vivo stability, including functional or metabolic reprogramming and epigenetic editing.

2.
Appl Microbiol Biotechnol ; 108(1): 179, 2024 Jan 27.
Article En | MEDLINE | ID: mdl-38280035

Several COVID-19 vaccines use adenovirus vectors to deliver the SARS-CoV-2 spike (S) protein. Immunization with these vaccines promotes immunity against the S protein, but against also the adenovirus itself. This could interfere with the entry of the vaccine into the cell, reducing its efficacy. Herein, we evaluate the efficiency of an adenovirus-vectored vaccine (chimpanzee ChAdOx1 adenovirus, AZD1222) in boosting the specific immunity compared to that induced by a recombinant receptor-binding domain (RBD)-based vaccine without viral vector. Mice immunized with the AZD1222 human vaccine were given a booster 6 months later, with either the homologous vaccine or a recombinant vaccine based on RBD of the delta variant, which was prevalent at the start of this study. A significant increase in anti-RBD antibody levels was observed in rRBD-boosted mice (31-61%) compared to those receiving two doses of AZD1222 (0%). Significantly higher rates of PepMix™- or RBD-elicited proliferation were also observed in IFNγ-producing CD4 and CD8 cells from mice boosted with one or two doses of RBD, respectively. The lower efficiency of the ChAdOx1-S vaccine in boosting specific immunity could be the result of a pre-existing anti-vector immunity, induced by increased levels of anti-adenovirus antibodies found both in mice and humans. Taken together, these results point to the importance of avoiding the recurrent use of the same adenovirus vector in individuals with immunity and memory against them. It also illustrates the disadvantages of ChAdOx1 adenovirus-vectored vaccine with respect to recombinant protein vaccines, which can be used without restriction in vaccine-booster programs. KEY POINTS: • ChAdOx1 adenovirus vaccine (AZD1222) may not be effective in boosting anti-SARS-CoV-2 immunity • A recombinant RBD protein vaccine is effective in boosting anti-SARS-CoV-2 immunity in mice • Antibodies elicited by the rRBD-delta vaccine persisted for up to 3 months in mice.


Adenovirus Vaccines , COVID-19 , Vaccines , Humans , Animals , Mice , Pan troglodytes , ChAdOx1 nCoV-19 , COVID-19 Vaccines/genetics , SARS-CoV-2 , COVID-19/prevention & control , Adenoviridae/genetics , Vaccination , Antibodies, Viral , Antibodies, Neutralizing
3.
Arch Med Res ; 55(2): 102960, 2024 Feb.
Article En | MEDLINE | ID: mdl-38290199

BACKGROUND: SARS-CoV2 induces flu-like symptoms that can rapidly progress to severe acute lung injury and even death. The virus also invades the central nervous system (CNS), causing neuroinflammation and death from central failure. Intravenous (IV) or oral dexamethasone (DXM) reduced 28 d mortality in patients who required supplemental oxygen compared to those who received conventional care alone. Through these routes, DMX fails to reach therapeutic levels in the CNS. In contrast, the intranasal (IN) route produces therapeutic levels of DXM in the CNS, even at low doses, with similar systemic bioavailability. AIMS: To compare IN vs. IV DXM treatment in hospitalized patients with COVID-19. METHODS: A controlled, multicenter, open-label trial. Patients with COVID-19 (69) were randomly assigned to receive IN-DXM (0.12 mg/kg for three days, followed by 0.6 mg/kg for up to seven days) or IV-DXM (6 mg/d for 10 d). The primary outcome was clinical improvement, as defined by the National Early Warning Score (NEWS) ordinal scale. The secondary outcome was death at 28 d between IV and IN patients. Effects of both treatments on biochemical and immunoinflammatory profiles were also recorded. RESULTS: Initially, no significant differences in clinical severity, biometrics, and immunoinflammatory parameters were found between both groups. The NEWS-2 score was reduced, in 23 IN-DXM treated patients, with no significant variations in the 46 IV-DXM treated ones. Ten IV-DXM-treated patients and only one IN-DXM patient died. CONCLUSIONS: IN-DMX reduced NEWS-2 and mortality more efficiently than IV-DXM, suggesting that IN is a more efficient route of DXM administration.


COVID-19 , Humans , SARS-CoV-2 , RNA, Viral , COVID-19 Drug Treatment , Dexamethasone/therapeutic use
4.
J Pharm Biomed Anal ; 235: 115680, 2023 Oct 25.
Article En | MEDLINE | ID: mdl-37634360

Biotherapeutics are complex molecules with therapeutic activity produced through biotechnology and/or genetic engineering. These medicines have clinical applications in diagnostic procedures and therapies for many disorders, including cancer, autoimmunity, and chronic degenerative diseases. Most biotherapeutics are expensive and sometimes unaffordable for low-income patients suffering from cancer or chronic illness. Biosimilars emerged in the 2000 s after patents of many innovative biotherapeutic products expired. The Biosimilar market is growing fast and demands reliable technologies for analyzing the physicochemical properties and bioactivity of products. A big challenge for biosimilar development is to prove comparable bioactivity, safety, efficacy, and toxicity profile as the innovator product. Bioactivity assessment can utilize different analytical techniques such as ELISA, flow cytometry, and surface plasmon resonance. Flow cytometry is a versatile analytical tool that can be used for the development of quantitative, reproducible, and accurate protocols suitable for routine evaluation of bioactivity in-vitro. Nevertheless, flow cytometry has been very scarcely used in comparability evaluation between biosimilar versus an originator product. Here, we review potential applications of flow cytometry to carry out functional bioassays of biotherapeutics or biosimilars.


Biosimilar Pharmaceuticals , Humans , Flow Cytometry , Biological Assay , Biotechnology , Enzyme-Linked Immunosorbent Assay
5.
Front Immunol ; 14: 1088039, 2023.
Article En | MEDLINE | ID: mdl-36855628

The transforming growth factor receptor III (TßRIII) is commonly recognized as a co-receptor that promotes the binding of TGFß family ligands to type I and type II receptors. Within the immune system, TßRIII regulates T cell development in the thymus and is differentially expressed through activation; however, its function in mature T cells is unclear. To begin addressing this question, we developed a conditional knock-out mouse with restricted TßRIII deletion in mature T cells, necessary because genomic deletion of TßRIII results in perinatal mortality. We determined that TßRIII null mice developed more severe autoimmune central nervous neuroinflammatory disease after immunization with myelin oligodendrocyte peptide (MOG35-55) than wild-type littermates. The increase in disease severity in TßRIII null mice was associated with expanded numbers of CNS infiltrating IFNγ+ CD4+ T cells and cells that co-express both IFNγ and IL-17 (IFNγ+/IL-17+), but not IL-17 alone expressing CD4 T cells compared to Tgfbr3fl/fl wild-type controls. This led us to speculate that TßRIII may be involved in regulating conversion of encephalitogenic Th17 to Th1. To directly address this, we generated encephalitogenic Th17 and Th1 cells from wild type and TßRIII null mice for passive transfer of EAE into naïve mice. Remarkably, Th17 encephalitogenic T cells from TßRIII null induced EAE of much greater severity and earlier in onset than those from wild-type mice. The severity of EAE induced by encephalitogenic wild-type and Tgfbr3fl/fl.dLcKCre Th1 cells were similar. Moreover, in vitro restimulation of in vivo primed Tgfbr3fl/fl.dLcKCre T cells, under Th17 but not Th1 polarizing conditions, resulted in a significant increase of IFNγ+ T cells. Altogether, our data indicate that TßRIII is a coreceptor that functions as a key checkpoint in controlling the pathogenicity of autoreactive T cells in neuroinflammation probably through regulating plasticity of Th17 T cells into pathogenic Th1 cells. Importantly, this is the first demonstration that TßRIII has an intrinsic role in T cells.


Encephalitis , Receptors, Transforming Growth Factor beta , Th17 Cells , Animals , Female , Mice , Pregnancy , Phosphorylation , Receptors, Transforming Growth Factor beta/genetics
6.
Front Immunol ; 14: 1062456, 2023.
Article En | MEDLINE | ID: mdl-36911743

The development of new strategies based on the use of Tr1 cells has taken relevance to induce long-term tolerance, especially in the context of allogeneic stem cell transplantation. Although Tr1 cells are currently identified by the co-expression of CD49b and LAG-3 and high production of interleukin 10 (IL-10), recent studies have shown the need for a more exhaustive characterization, including co-inhibitory and chemokines receptors expression, to ensure bona fide Tr1 cells to be used as cell therapy in solid organ transplantation. Moreover, the proinflammatory environment induced by the allograft could affect the suppressive function of Treg cells, therefore stability of Tr1 cells needs to be further investigated. Here, we establish a new protocol that allows long-term in vitro expansion of highly purified expanded allospecific Tr1 (Exp-allo Tr1). Our expanded Tr1 cell population becomes highly enriched in IL-10 producers (> 90%) and maintains high expression of CD49b and LAG-3, as well as the co-inhibitory receptors PD-1, CTLA-4, TIM-3, TIGIT and CD39. Most importantly, high dimensional analysis of Exp-allo Tr1 demonstrated a specific expression profile that distinguishes them from activated conventional T cells (T conv), showing overexpression of IL-10, CD39, CTLA-4 and LAG-3. On the other hand, Exp-allo Tr1 expressed a chemokine receptor profile relevant for allograft homing and tolerance induction including CCR2, CCR4, CCR5 and CXCR3, but lower levels of CCR7. Interestingly, Exp-allo Tr1 efficiently suppressed allospecific but not third-party T cell responses even after being expanded in the presence of proinflammatory cytokines for two extra weeks, supporting their functional stability. In summary, we demonstrate for the first time that highly purified allospecific Tr1 (Allo Tr1) cells can be efficiently expanded maintaining a stable phenotype and suppressive function with homing potential to the allograft, so they may be considered as promising therapeutic tools for solid organ transplantation.


Hematopoietic Stem Cell Transplantation , Organ Transplantation , T-Lymphocytes, Regulatory/metabolism , Interleukin-10/metabolism , CTLA-4 Antigen/metabolism , Integrin alpha2/metabolism
7.
PLoS One ; 17(4): e0266827, 2022.
Article En | MEDLINE | ID: mdl-35472214

Growing evidence demonstrates a strong correlation between obesity and an increased risk of breast cancer, although the mechanisms involved have not been completely elucidated. Some reports have described a crosstalk between adipocytes, cancer cells, and immune cells within the tumor microenvironment, however, it is currently unknown whether obesity can promote tumor growth by inducing systemic alterations of the immune cell homeostasis in peripheral lymphoid organs and adipose tissue. Here, we used the E0771 breast cancer cell line in a mouse model of diet-induced obesity to analyze the immune subpopulations present in the tumors, visceral adipose tissue (VAT), and spleen of lean and obese mice. Our results showed a significant reduction in the frequency of infiltrating CD8+ T cells and a decreased M1/M2 macrophage ratio, indicative of the compromised anti-tumoral immune response reported in obesity. Despite not finding differences in the percentage or numbers of intratumoral Tregs, phenotypic analysis showed that they were enriched in CD39+, PD-1+ and CCR8+ cells, compared to the draining lymph nodes, confirming the highly immunosuppressive profile of infiltrating Tregs reported in established tumors. Analysis of peripheral T lymphocytes showed that tumor development in obese mice was associated to a significant increase in the percentage of peripheral Tregs, which supports the systemic immunosuppressive effect caused by the tumor. Interestingly, evaluation of immune subpopulations in the VAT showed that the characteristic increase in the M1/M2 macrophage ratio reported in obesity, was completely reversed in tumor-bearing mice, resembling the M2-polarized profile found in the microenvironment of the growing tumor. Importantly, VAT Tregs, which are commonly decreased in obese mice, were significantly increased in the presence of breast tumors and displayed significantly higher levels of Foxp3, indicating a regulatory feedback mechanism triggered by tumor growth. Altogether, our results identify a complex reciprocal relationship between adipocytes, immune cells, and the tumor, which may modulate the immune macroenvironment that promotes breast cancer development in obesity.


Neoplasms , T-Lymphocytes, Regulatory , Animals , CD8-Positive T-Lymphocytes , Mice , Obesity/complications , Tumor Microenvironment
8.
Front Immunol ; 12: 686530, 2021.
Article En | MEDLINE | ID: mdl-34777330

The adoptive transfer of alloantigen-specific regulatory T cells (alloTregs) has been proposed as a therapeutic alternative in kidney transplant recipients to the use of lifelong immunosuppressive drugs that cause serious side effects. However, the clinical application of alloTregs has been limited due to their low frequency in peripheral blood and the scarce development of efficient protocols to ensure their purity, expansion, and stability. Here, we describe a new experimental protocol that allows the long-term expansion of highly purified allospecific natural Tregs (nTregs) from both healthy controls and chronic kidney disease (CKD) patients, which maintain their phenotype and suppressive function under inflammatory conditions. Firstly, we co-cultured CellTrace Violet (CTV)-labeled Tregs from CKD patients or healthy individuals with allogeneic monocyte-derived dendritic cells in the presence of interleukin 2 (IL-2) and retinoic acid. Then, proliferating CD4+CD25hiCTV- Tregs (allospecific) were sorted by fluorescence-activated cell sorting (FACS) and polyclonally expanded with anti-CD3/CD28-coated beads in the presence of transforming growth factor beta (TGF-ß), IL-2, and rapamycin. After 4 weeks, alloTregs were expanded up to 2,300 times the initial numbers with a purity of >95% (CD4+CD25hiFOXP3+). The resulting allospecific Tregs showed high expressions of CTLA-4, LAG-3, and CD39, indicative of a highly suppressive phenotype. Accordingly, expanded alloTregs efficiently suppressed T-cell proliferation in an antigen-specific manner, even in the presence of inflammatory cytokines (IFN-γ, IL-4, IL-6, or TNF-α). Unexpectedly, the long-term expansion resulted in an increased methylation of the specific demethylated region of Foxp3. Interestingly, alloTregs from both normal individuals and CKD patients maintained their immunosuppressive phenotype and function after being expanded for two additional weeks under an inflammatory microenvironment. Finally, phenotypic and functional evaluation of cryopreserved alloTregs demonstrated the feasibility of long-term storage and supports the potential use of this cellular product for personalized Treg therapy in transplanted patients.


Cytokines/metabolism , Inflammation Mediators/metabolism , Isoantigens/immunology , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Biomarkers , Cellular Microenvironment/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Susceptibility , Flow Cytometry , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Phenotype , Renal Insufficiency, Chronic/diagnosis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
9.
Int J Biol Macromol ; 187: 76-90, 2021 Sep 30.
Article En | MEDLINE | ID: mdl-34280449

Lupin γ-conglutin beneficially modulates glycemia, but whether it protects against oxidative and lipotoxic damage remains unknown. Here, we studied the effects of γ-conglutin on cell death provoked by hydrogen peroxide and palmitate in HepG2 hepatocytes and insulin-producing MIN6 cells, and if a modulation of mitochondrial potential and reactive oxygen species (ROS) levels was involved. We also investigated how γ-conglutin influences insulin secretion and electrical activity of ß-cells. The increased apoptosis of HepG2 cells exposed to hydrogen peroxide was prevented by γ-conglutin, and the viability and ROS content in γ-conglutin-treated cells was similar to that of non-exposed cells. Additionally, γ-conglutin partially protected MIN6 cells against hydrogen peroxide-induced death. This was associated with a marked reduction in ROS. No significant changes were found in the mitochondrial potential of γ-conglutin-treated cells. Besides, we observed a partial protection against lipotoxicity only in hepatocytes. Unexpectedly, we found a transient inhibition of insulin secretion, plasma membrane hyperpolarization, and higher KATP channel currents in ß-cells treated with γ-conglutin. Our data show that γ-conglutin protects against cell death induced by oxidative stress or lipotoxicity by decreasing ROS and might also indicate that γ-conglutin promotes a ß-cell rest, which could be useful for preventing ß-cell exhaustion in chronic hyperglycemia.


Insulin Secretion/drug effects , Insulin-Secreting Cells/metabolism , Lupinus/chemistry , Membrane Potentials/drug effects , Oxidative Stress/drug effects , P-type ATPases/metabolism , Plant Proteins , Animals , Cell Death/drug effects , Hep G2 Cells , Humans , Hydrogen Peroxide , Mice , Plant Proteins/chemistry , Plant Proteins/pharmacology
10.
Methods Mol Biol ; 2174: 171-191, 2021.
Article En | MEDLINE | ID: mdl-32813250

The study of tumor exosomes has gained relevance in the last decades due to their potential use for therapeutic and diagnostic application. Although there is extensive knowledge of exosome biology, some biological samples like tumor-derived exosomes have been difficult to characterize due to their complexity and heterogeneity. This distinctive feature makes difficult the identification of specific exosome subpopulations with a shared molecular signature that could allow for targeting of exosomes with therapeutic and diagnostic potential use in cancer patients. Nanoscale flow cytometry has lately emerged as an alternative tool that can be adapted to the study of nanoparticles, such as exosomes. However, the physicochemical properties of these particles are an important issue to consider as nanoparticles need the application of specific settings which differ from those used in conventional flow cytometry of cells. Therefore, in the last few years, one of the main aims has been the optimization of technical and experimental protocols to improve exosome analysis. In this chapter, we discuss several aspects of cytometric systems with a special emphasis in technical considerations of samples and equipment.


Exosomes/chemistry , Exosomes/pathology , Flow Cytometry/methods , Neoplasms/pathology , Calibration , Centrifugation, Density Gradient/methods , Chromatography, Gel/methods , Flow Cytometry/instrumentation , Humans , Nanotechnology/instrumentation , Nanotechnology/methods , Prognosis , Ultracentrifugation/methods
11.
Methods Mol Biol ; 2174: 219-244, 2021.
Article En | MEDLINE | ID: mdl-32813253

The use of immunotherapy as an alternative treatment for cancer patients has become of great interest in the scientific community as it is required to overcome many of the currently unsolved problems such as tumor escape, immunosuppression and unwanted unspecific toxicity. The use of chimeric antigen receptor T cells has been a very successful strategy in some hematologic malignancies. However, the application of CAR T cells has been limited to solid tumors, and this has aimed the development of new generation of CARs with enhanced effectivity and specificity. Here, we review the state of the art of CAR T cell therapy with special emphasis on the current challenges and opportunities.


Cell Transplantation/adverse effects , Cell Transplantation/methods , Neoplasms/therapy , Receptors, Chimeric Antigen , T-Lymphocytes/physiology , Genes, Transgenic, Suicide , Hematologic Neoplasms/therapy , Humans , Receptors, Chimeric Antigen/chemistry , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/cytology
12.
Front Immunol ; 11: 375, 2020.
Article En | MEDLINE | ID: mdl-32300340

Regulatory T cells play an important role in the control of autoimmune diseases and maintenance of tolerance. In the context of transplantation, regulatory T cells (Tregs) have been proposed as new therapeutic tools that may induce allospecific tolerance toward the graft, avoiding the side effects induced by generalized immunosuppressors. Although most clinical trials are based on the use of thymic Tregs in adoptive therapy, some reports suggest the potential use of in vitro induced Tregs (iTregs), based on their functional stability under inflammatory conditions, indicating an advantage in a setting of allograft rejection. The aim of this work was to generate and expand large numbers of allospecific Tregs that maintain stable suppressive function in the presence of pro-inflammatory cytokines. Dendritic cells were derived from monocytes isolated from healthy donors and were co-cultured with CTV-labeled naïve T cells from unrelated individuals, in the presence of TGF-ß1, IL-2, and retinoic acid. After 7 days of co-culture, proliferating CD4+CD25++CTV- cells (allospecific iTregs) were sorted and polyclonally expanded for 6 weeks in the presence of TGF-ß1, IL-2, and rapamycin. After 6 weeks of polyclonal activation, iTregs were expanded 230,000 times, giving rise to 4,600 million allospecific iTregs. Allospecific iTregs were able to specifically suppress the proliferation of autologous CD4+ and CD8+ T cells in response to the allo-MoDCs used for iTreg generation, but not to third-party allo-MoDCs. Importantly, 88.5% of the expanded cells were CD4+CD25+FOXP3+, expressed high levels of CCR4 and CXCR3, and maintained their phenotype and suppressive function in the presence of TNF-α and IL-6. Finally, analysis of the methylation status of the FOXP3 TSDR locus demonstrated a 40% demethylation in the purified allospecific iTreg, prior to the polyclonal expansion. Interestingly, the phenotype and suppressive activity of expanded allospecific iTregs were maintained after 6 weeks of expansion, despite an increase in the methylation status of the FOXP3 TSDR. In conclusion, this is the first report that demonstrates a large-scale generation of allospecific iTregs that preserve a stable phenotype and suppressor function in the presence of pro-inflammatory cytokines and pave the way for adoptive cell therapy with iTregs in transplanted patients.


Allogeneic Cells/immunology , Cell Culture Techniques/methods , Immunotherapy, Adoptive/methods , T-Lymphocytes, Regulatory/immunology , Allogeneic Cells/cytology , Humans , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/transplantation
13.
PLoS One ; 14(9): e0222301, 2019.
Article En | MEDLINE | ID: mdl-31491023

The role of CD5 as a regulator of T cell signaling and tolerance is well recognized. Recent data show expression of CD5 on different subtypes of human dendritic cells, however its functional relevance in modulating DC mediated responses remains poorly understood. In this study, we show CD5 is expressed on CD11c+ DC from murine thymus, lymph node, spleen, skin and lung. Although the development of DC subpopulations in CD5-/- mice was normal, CD5-deficient DC produced significantly higher levels of IL-12 than wild type DC in response to LPS. CD5-/- DC, in comparison to CD5+/+ DC, enhanced the activation of CD4+ and CD8+ T cells in vitro and in vivo and induced significantly higher production of IL-2 and IFN-gamma by T cells. Consequently, CD5-/- DC were significantly more potent than wild type DC in the induction of anti-tumor immunity and contact hypersensitivity responses in mice. Restoration of CD5 expression in CD5-/- DC reduced IL-12 production and inhibited their capacity to stimulate T cells. Collectively, these data demonstrate that the specific expression of CD5 on DC inhibits the production of inflammatory cytokines and has a regulatory effect on their activity to stimulate T cells and induce immune responses. This study reveals a previously unrecognized regulatory role for CD5 on DC and provides novel insights into mechanisms for DC biology in immune responses.


CD4-Positive T-Lymphocytes/metabolism , CD5 Antigens/metabolism , CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/metabolism , Lymphocyte Activation/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD5 Antigens/genetics , CD8-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Female , Male , Mice , Mice, Transgenic
14.
Cancers (Basel) ; 11(6)2019 May 30.
Article En | MEDLINE | ID: mdl-31151160

Hypoxia and the accumulation of hypoxia-inducible factors (HIFs) in tumors have been associated with therapeutic resistance and with autophagy establishment. We examined the effects of stable knockdown of HIF-1α or HIF-2α expression on autophagy and drug resistance in colon cancer cells. We found that under normoxic conditions, malignant cells exhibit increased basal levels of autophagy, compared with non-malignant cells, in addition to the previously reported coexpression of HIF-1α and HIF-2α. Knockdown of HIF-1α or HIF-2α expression resulted in increased autophagic and apoptotic cell death, indicating that the survival of cells is HIF-dependent. Cytotoxic-induced cell death was significantly increased by knockdown of HIFs but not by autophagy inhibition. Strikingly, although malignancy-resistant cells were sensitized to death by nutrient stress, the combination with HIF-2α depletion, but not with HIF-1α depletion, induced severe cell death. Oxidative stress levels were significantly increased as a result of HIF-2α specific inhibition or silencing suggesting that this may contribute to sensitize cells to death. The in vitro results were confirmed in vivo using a xenograft mouse model. We found that coordinated autophagy and mTOR inhibition enhanced cell death and induced tumor remission only in HIF-2α-silenced cells. Finally, using a specific HIF-2α inhibitor alone or in combination with drugs in patient-derived primary colon cancer cells, overcame their resistance to 5-FU or CCI-779, thus emphasizing the crucial role played by HIF-2α in promoting resistance and cell survival.

15.
J Leukoc Biol ; 105(5): 891-904, 2019 05.
Article En | MEDLINE | ID: mdl-30676652

Since its discovery, over 30 years ago, CD5 has been used as a marker to identify T cells, B1-a cells, and B cell chronic lymphocytic leukemia cells. Throughout the years, many studies have described the functional relevance of CD5 as a modulator of T and B cell receptor signaling. However, it has not been until recent years that CD5 has emerged as a functional receptor in other areas of the immune system. Here, we review some of the most important aspects of CD5 as a modulator of TCR and BCR signaling, cell survival receptor both in T and B cells during health and disease, as well as the newly discovered roles of this receptor in thymocyte selection, T cell effector differentiation, and immune tolerance. CD5 was found to promote T cell survival by protecting autoreactive T cell from activation-induced cell death, to promote de novo induction of regulatory T cells in the periphery, to modulate Th17 and Th2 differentiation, and to modulate immune responses by modulating dendritic cell functions. CD5 is overexpressed in Tregs and Bregs, which are fundamental to maintain immune homeostasis. The newly established roles of CD5 in modulating different aspects of immune responses identify this receptor as an immune checkpoint modulator, and therefore it could be used as a target for immune intervention in different pathologies such as cancer, autoimmune diseases or infections.


Autoimmune Diseases/genetics , B-Lymphocytes, Regulatory/immunology , CD5 Antigens/genetics , Communicable Diseases/genetics , Neoplasms/genetics , T-Lymphocytes, Regulatory/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , B-Lymphocytes, Regulatory/pathology , CD5 Antigens/immunology , Cell Differentiation , Cell Survival , Communicable Diseases/immunology , Communicable Diseases/pathology , Dendritic Cells/immunology , Dendritic Cells/pathology , Gene Expression Regulation , Homeostasis/genetics , Homeostasis/immunology , Humans , Immune Tolerance , Lymphocyte Activation , Neoplasms/immunology , Neoplasms/pathology , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Signal Transduction , T-Lymphocytes, Regulatory/pathology , Th17 Cells/immunology , Th17 Cells/pathology
16.
FEBS Open Bio ; 9(1): 137-147, 2019 01.
Article En | MEDLINE | ID: mdl-30652081

We have previously reported that the absence of inhibins results in impaired dendritic cell (DC) maturation and function, leading to decreased T cell activation and diminished delayed-type hypersensitivity responses. Here, we investigated the role of inhibins in peripheral regulatory T cell (Treg) induction in vitro and in vivo. Inhibin deficient (Inhα-/-) mice showed an increased percentage of peripherally induced Tregs in colonic lamina propria and mesenteric lymph nodes, compared to Inhα+/+ mice, which correlated with increased expression of PD-L1 in CD103+ and CD8α+ DCs. Lipopolysaccharide-stimulated bone marrow-derived and ex vivo spleen- and lymph node-purified CD11c+ Inhα-/- DCs induced higher Tregs in vitro. Moreover, in vivo anti-DEC205-ovalbumin (OVA) DC targeting of mice with adoptively transferred OVA-specific T cells showed enhanced induced peripheral Treg conversion in Inhα-/- mice. These data identify inhibins as key regulators of peripheral T cell tolerance.


Dendritic Cells/immunology , Dendritic Cells/metabolism , Inhibins/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Cells, Cultured , Dendritic Cells/cytology , Inhibins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout
17.
J Anim Sci ; 97(2): 644-656, 2019 Feb 01.
Article En | MEDLINE | ID: mdl-30517644

Tilmicosin is an antimicrobial agent used to treat intramammary infections against Staphylococcus aureus and has clinical anti-inflammatory effects. However, the mechanism by which it modulates the inflammatory process in the mammary gland is unknown. We evaluated the effect of tilmicosin treatment on the modulation of the mammary innate immune response after S. aureus infection and its effect on casein production in mammary epithelial cells. To achieve this goal, we used immortalized mammary epithelial cells (MAC-T), pretreated for 12 h or treated with tilmicosin after infection with S. aureus (ATCC 27543). Our data showed that tilmicosin decreases intracellular infection (P < 0.01) and had a protective effect on MAC-T reducing apoptosis after infection by 80% (P < 0.01). Furthermore, tilmicosin reduced reactive oxygen species (ROS) (P < 0.01), IL-1ß (P < 0.01), IL-6 (P < 0.01), and TNF-α (P < 0.05) production. In an attempt to investigate the signaling pathways involved in the immunomodulatory effect of tilmicosin, mitogen-activated protein kinase (MAPK) phosphorylation was measured by fluorescent-activated cell sorting. Pretreatment with tilmicosin increased ERK1/2 (P < 0.05) but decreased P38 phosphorylation (P < 0.01). In addition, the anti-inflammatory effect of tilmicosin helped to preserve casein synthesis in mammary epithelial cells (P < 0.01). This result indicates that tilmicosin could be an effective modulator inflammation in the mammary gland. Through regulation of MAPK phosphorylation, ROS production and pro-inflammatory cytokine secretion tilmicosin can provide protection from cellular damage due to S. aureus infection and help to maintain normal physiological functions of the bovine mammary epithelial cell.


Anti-Bacterial Agents/pharmacology , Caseins/metabolism , Immunity, Innate/drug effects , Mastitis, Bovine/drug therapy , Staphylococcal Infections/veterinary , Staphylococcus aureus/drug effects , Tylosin/analogs & derivatives , Alveolar Epithelial Cells/metabolism , Animals , Cattle , Cytokines/metabolism , Female , Mammary Glands, Animal/metabolism , Mastitis, Bovine/microbiology , Mitogen-Activated Protein Kinases/metabolism , Signal Transduction/drug effects , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Tylosin/pharmacology
18.
Front Immunol ; 9: 2212, 2018.
Article En | MEDLINE | ID: mdl-30319653

CD4+ T cells are major players in the immune response against several diseases; including AIDS, leishmaniasis, tuberculosis, influenza and cancer. Their activation has been successfully achieved by administering antigen coupled with antibodies, against DC-specific receptors in combination with adjuvants. Unfortunately, most of the adjuvants used so far in experimental models are unsuitable for human use. Therefore, human DC-targeted vaccination awaits the description of potent, yet nontoxic adjuvants. The nontoxic cholera B subunit (CTB) can be safely used in humans and it has the potential to activate CD4+ T cell responses. However, it remains unclear whether CTB can promote DC activation and can act as an adjuvant for DC-targeted antigens. Here, we evaluated the CTB's capacity to activate DCs and CD4+ T cell responses, and to generate long-lasting protective immunity. Intradermal (i.d.) administration of CTB promoted late and prolonged activation and accumulation of skin and lymphoid-resident DCs. When CTB was co-administered with anti-DEC205-OVA, it promoted CD4+ T cell expansion, differentiation, and infiltration to peripheral nonlymphoid tissues, i.e., the skin, lungs and intestine. Indeed, CTB promoted a polyfunctional CD4+ T cell response, including the priming of Th1 and Th17 cells, as well as resident memory T (RM) cell differentiation in peripheral nonlymphoid tissues. It is worth noting that CTB together with a DC-targeted antigen promoted local and systemic protection against experimental melanoma and murine rotavirus. We conclude that CTB administered i.d. can be used as an adjuvant to DC-targeted antigens for the induction of broad CD4+ T cell responses as well as for promoting long-lasting protective immunity.


Adjuvants, Immunologic/administration & dosage , Cholera Toxin/administration & dosage , Dendritic Cells/immunology , Lectins, C-Type/antagonists & inhibitors , Receptors, Cell Surface/antagonists & inhibitors , Vaccination/methods , Animals , Antigens, CD/immunology , Cell Line, Tumor/transplantation , Disease Models, Animal , Female , Humans , Injections, Intradermal , Lectins, C-Type/immunology , Lymphocyte Activation/immunology , Male , Melanoma/immunology , Melanoma/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Minor Histocompatibility Antigens/immunology , Receptors, Cell Surface/immunology , Rotavirus/immunology , Rotavirus Infections/immunology , Rotavirus Infections/prevention & control , Rotavirus Infections/virology , Th1 Cells/immunology , Th17 Cells/immunology , Treatment Outcome
19.
Biochem Biophys Res Commun ; 494(1-2): 82-87, 2017 12 09.
Article En | MEDLINE | ID: mdl-29050936

TGF-ß type III receptor (TßRIII) is a co-receptor for TGFß family members required for high-affinity binding of these ligands to their receptors, potentiating their cellular functions. TGF-ßs, Bone Morphogenetic Proteins (BMP2/4) and Inhibins/Activins regulate different checkpoints during T cell differentiation. We have previously reported that TßRIII modulates T cell development by protecting developing thymocytes from apoptosis, however the role of this co-receptor in peripheral lymphocytes still remains elusive. Here we describe a detailed characterization of TßRIII expression in murine and human lymphocyte subpopulations demonstrating that this co-receptor is significantly expressed in T but not B lymphocytes and among them, preferentially expressed on naïve and central memory T cells. TßRIII was upregulated after TCR stimulation, in parallel to other early activation markers. In contrast, natural and induced Tregs downregulated TßRIII in association with FoxP3 upregulation. Finally, anti-TßRIII blocking experiments demonstrated that TßRIII promotes TGFß-dependent iTreg conversion in vitro, and suggest that this co-receptor may be involved in modulating peripheral T cell tolerance and could be considered as a potential target to boost T cell immune responses.


Proteoglycans/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, Transforming Growth Factor beta/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Down-Regulation , Forkhead Transcription Factors/metabolism , Humans , Immunologic Memory , In Vitro Techniques , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proteoglycans/antagonists & inhibitors , Proteoglycans/genetics , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , T-Lymphocytes, Regulatory/classification , T-Lymphocytes, Regulatory/metabolism , Up-Regulation
20.
Front Immunol ; 8: 219, 2017.
Article En | MEDLINE | ID: mdl-28316600

Regulatory T cells (Tregs) are considered key players in the prevention of allograft rejection in transplanted patients. Belatacept (BLT) is an effective alternative to calcineurin inhibitors that appears to preserve graft survival and function; however, the impact of this drug in the homeostasis of Tregs in transplanted patients remains controversial. Here, we analyzed the phenotype, function, and the epigenetic status of the Treg-specific demethylated region (TSDR) in FOXP3 of circulating Tregs from long-term kidney transplant patients under BLT or Cyclosporine A treatment. We found a significant reduction in the proportion of CD4+CD25hiCD127lo/-FOXP3+ T cells in all patients compared to healthy individual (controls). Interestingly, only BLT-treated patients displayed an enrichment of the CD45RA+ "naïve" Tregs, while the expression of Helios, a marker used to identify stable FOXP3+ thymic Tregs remained unaffected. Functional analysis demonstrated that Tregs from transplanted patients displayed a significant reduction in their suppressive capacity compared to Tregs from controls, which is associated with decreased levels of FOXP3 and CD25. Analysis of the methylation status of the FOXP3 gene showed that BLT treatment results in methylation of CpG islands within the TSDR, which could be associated with the impaired Treg suppression function. Our data indicate that analysis of circulating Tregs cannot be used as a marker for assessing tolerance toward the allograft in long-term kidney transplant patients. Trial registration number IM103008.

...