Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters

Affiliation country
Publication year range
1.
Cancer Immunol Immunother ; 72(8): 2687-2700, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37081323

ABSTRACT

Adaptive NK cells constitute an NK cell subpopulation, which expands after human cytomegalovirus (HCMV) infection. This subpopulation has stronger production of cytokines after CD16 stimulation, longer life and persistence than conventional NK cells and are, therefore, interesting tools for cancer immunotherapy. Since there is limited information on adaptive NK cells in cancer patients, we described this population phenotypically and functionally, by flow cytometry, in the context of HER2 + breast cancer (BC) directed therapy. We assessed HCMV status in 78 patients with BC. We found that, similarly to healthy donors (HD), a high proportion of BC patients were HCMV-positive, and nearly 72% of them had an adaptive NK cell subpopulation characterized by the loss of FcεRIγ intracellular adaptor protein or the presence of NKG2C receptor. However, in BC patients, FcεRIγ- and NKG2C + NK cell populations overlapped to a lesser extent than in HD. Otherwise, no profound phenotypic differences were found between BC patients and HD. Although FcεRIγ- or NKG2C + NK cell subsets from BC patients produced more IFN-γ than their FcεRIγ + or NKG2C- NK cell counterparts, IFN-γ production increased only when NK cells simultaneously expressed FcεRIγ- and NKG2C + , whereas in HD the presence of NKG2C marker was sufficient to display greater functionality. Furthermore, in a group of patients treated with chemotherapy and Trastuzumab plus Pertuzumab, FcεRIγ-NKG2C + and FcεRIγ-NKG2C- NK cells retained greater functionality after treatment than FcεRIγ + NKG2C- NK cells. These results suggest that the presence or magnitude of adaptive NK cell subsets might serve as a key determinant for therapeutic approaches based on antibodies directed against tumor antigens.


Subject(s)
Breast Neoplasms , Cytomegalovirus Infections , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cytomegalovirus , Killer Cells, Natural , Cytokines , NK Cell Lectin-Like Receptor Subfamily C/metabolism
2.
Immunology ; 154(2): 261-273, 2018 06.
Article in English | MEDLINE | ID: mdl-29247515

ABSTRACT

Deep characterization of the frequencies, phenotypes and functionalities of liver and peripheral blood natural killer (NK), natural killer T (NKT) and T cells from healthy individuals is an essential step to further interpret changes in liver diseases. These data indicate that CCR7, a chemokine essential for cell migration through lymphoid organs, is almost absent in liver NK and T cells. CD56bright NK cells, which represent half of liver NK cells, showed lower expression of the inhibitory molecule NKG2A and an increased frequency of the activation marker NKp44. By contrast, a decrease of CD16 expression with a potential decreased capacity to perform antibody-dependent cellular cytotoxicity was the main difference between liver and peripheral blood CD56dim NK cells. Liver T cells with an effector memory or terminally differentiated phenotype showed an increased frequency of MAIT cells,T-cell receptor-γδ (TCR-γδ) T cells and TCR-αß CD8+ cells, with few naive T cells. Most liver NK and T cells expressed the homing markers CD161 and CD244. Liver T cells revealed a unique expression pattern of killer cell immunoglobulin-like receptors (KIR) receptors, with increased degranulation ability and higher secretion of interferon-γ. Hence, the liver possesses a large amount of memory and terminally differentiated CD8+ cells with a unique expression pattern of KIR activating receptors that have a potent functional capacity as well as a reduced amount of CCR7, which are unable to migrate to regional lymph nodes. These results are consistent with previous studies showing that liver T (and also NK) cells likely remain and die in the liver.


Subject(s)
Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Liver/immunology , Liver/metabolism , Phenotype , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Antigens, Surface/metabolism , Biomarkers , Cell Differentiation/immunology , Humans , Immunophenotyping , Killer Cells, Natural/cytology , Lymphocyte Count , Receptors, Natural Killer Cell/metabolism , T-Lymphocyte Subsets/cytology
3.
J Immunol ; 196(8): 3287-96, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26951799

ABSTRACT

Obesity-induced inflammation is conducted by a metabolic pathway, which eventually causes activation of specialized immune cells and leads to an unresolved inflammatory response within the tissue. For this reason, it is critically important to determine how hypertrophic fat tissue alters T cell balance to drive inflammation. In this study, we identify the purinergic signaling as a novel mechanism driving the adaptive Th17 response in human visceral adipose tissue (VAT) of metabolically unhealthy obese patients. We demonstrate that ATP acting via the P2X7 receptor pathway promotes a Th17 polarizing microenvironment with high levels of IL-1ß, IL-6, and IL-17 in VAT explants from lean donors. Moreover, in vitro blockade of the P2X7 receptor abrogates the levels of these cytokines. These findings are consistent with a greater frequency of Th17 cells in tissue from metabolically unhealthy obese donors, revealed not only by the presence of a baseline Th17-promoting milieu, but also by the higher expression of steadily recognized Th17 markers, such as RORC, IL-17 cytokine, and IL-23R, in comparison with metabolically healthy obese and lean donors. In addition, we demonstrate that CD39 expression on CD4(+)effector T cells represents a novel Th17 marker in the inflamed VAT, which also confers protection against ATP-induced cell death. The manipulation of the purinergic signaling might represent a new therapeutic target to shift the CD4(+)T cell balance under inflammatory conditions.


Subject(s)
Adenosine Triphosphate/metabolism , Intra-Abdominal Fat/immunology , Obesity/immunology , Receptors, Purinergic P2X7/metabolism , Th17 Cells/immunology , Adult , Antigens, CD/biosynthesis , Apoptosis/physiology , Apyrase/biosynthesis , Cellular Microenvironment/immunology , Female , Humans , Inflammation/immunology , Interleukin-17/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Intra-Abdominal Fat/cytology , Intra-Abdominal Fat/pathology , Male , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Obesity/pathology , Receptors, Interleukin/metabolism , Th17 Cells/metabolism
4.
Mol Biol Rep ; 45(5): 987-1000, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30003508

ABSTRACT

Historical records suggest that Chiriguano tribe is the result of a genetic admixture event. The process involved the arrival of Guaraní tribesmen descending from Amazonian region of Brazil along with groups of Arawak origin that inhabited the foothill plains of Bolivia. Later they arrived in Argentina at the beginning of the twentieth century. Aiming to test the historical records, we analysed a set of 46 samples collected at San Ramon de la Nueva Orán, Province of Salta, Argentina. A wide set of uni- and biparentally transmitted genetic markers were analysed, including 23 autosomal STRs; 46 AIM-DIPs and 24 AIM-SNPs all located at diverse autosomal chromosome locations; 23 Y-STRs and the entire mtDNA D-Loop sequence. Ancestry informative markers allowed for the detection of a strong Native American component in the genomes (> 94%), while all mtDNA haplotypes showed Native American characteristic motives, and 93% of Y-haplotypes belonged to the Q1a3a Y-haplogroup. The analysis of mitochondrial haplotypes and Y chromosome, although they did not match other populations, revealed a relationship between the Chiriguano and other groups of Guaraní and Arawak origin inhabiting Brazil and Bolivia, confirming, at least in part, the historical records describing the origins of Chiriguano tribal settlements in northwestern Argentina.


Subject(s)
Ethnicity/genetics , Genetics, Population/methods , Indians, South American/genetics , Argentina , Bolivia , Brazil , Chromosomes, Human, Y/genetics , DNA, Mitochondrial/genetics , Female , Genetic Markers/genetics , Haplotypes , Humans , Male , Polymorphism, Single Nucleotide/genetics
5.
Eur J Immunol ; 45(5): 1560-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25726929

ABSTRACT

Clinical studies suggest that triple negative breast cancer (TNBC) patients with epidermal growth factor receptor (EGFR)-expressing tumors could benefit from therapy with Cetuximab, which targets EGFR. NK cells are the primary effectors of antibody (Ab)-dependent cell-mediated cytotoxicity (ADCC) and thus play a role in Ab-based therapies. We have previously described diminished levels of Cetuximab-mediated ADCC in vitro in patients with advanced breast cancer. Here, we investigated the potential causes of this NK-cell functional deficiency. We characterized NK-cell activating/inhibitory receptors in the peripheral blood of breast cancer patients and found CD85j inhibitory receptor overexpression. The capacity of NK cells to perform Cetuximab-triggered ADCC against TNBC cells correlated inversely with CD85j expression, even in the presence of the stimulatory cytokines IL-2 or IL-15. Hence, patients expressing high levels of CD85j had an impaired ability to lyse TNBC cells in the presence of Cetuximab. We also found that CD85j overexpression was associated with HLA-I and soluble HLA-G expression by tumors. A CD85j functional blockade with a CD85j antagonist Ab restored ADCC levels in breast cancer patients and reverted this negative effect. Our data suggest that strategies that overcome the hurdles of immune activation could improve Cetuximab clinical efficacy.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antibody-Dependent Cell Cytotoxicity , Antigens, CD/metabolism , Killer Cells, Natural/immunology , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/metabolism , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/therapy , Adult , Antineoplastic Agents/pharmacology , Case-Control Studies , Cetuximab , ErbB Receptors/antagonists & inhibitors , Female , HLA Antigens/metabolism , HLA-G Antigens/metabolism , Humans , K562 Cells , Leukocyte Immunoglobulin-like Receptor B1 , Middle Aged , Young Adult
6.
Cancer Immunol Immunother ; 65(5): 551-62, 2016 May.
Article in English | MEDLINE | ID: mdl-26969612

ABSTRACT

The expression of N-glycolyl-monosialodihexosyl-ganglioside (NGcGM3) in humans is restricted to cancer cells; therefore, it is a tumor antigen. There are measurable quantities of circulating anti-NGcGM3 antibodies (aNGcGM3 Abs) in human serum. Interestingly, some people have circulating Ag-specific immunoglobulins G (IgGs) that are capable of complement mediated cytotoxicity against NGcGM3 positive cells, which is relevant for tumor surveillance. In light of the chemical nature of Ag, we postulated it as a candidate ligand for CD1d. Furthermore, we hypothesize that the immune mechanism involved in the generation of these Abs entails cross talk between B lymphocytes (Bc) and invariant natural killer T cells (iNKT). Combining cellular techniques, such as flow cytometry and biochemical assays, we demonstrated that CD1d binds to NGcGM3 and that human Bc present NGcGM3 in a CD1d context according to two alternative strategies. We also showed that paraformaldehyde treatment of cells expressing CD1d affects the presentation. Finally, by co-culturing primary human Bc with iNKT and measuring Ki-67 expression, we detected a reproducible increment in the proliferation of the iNKT population when Ag was on the medium. Our findings identify a novel, endogenous, human CD1d ligand, which is sufficiently competent to stimulate iNKT. We postulate that CD1d-restricted Bc presentation of NGcGM3 drives effective iNKT activation, an immunological mechanism that has not been previously described for humans, which may contribute to understanding aNGcGM3 occurrence.


Subject(s)
Antigens, CD1d/immunology , B-Lymphocytes/immunology , Cell Communication/immunology , G(M3) Ganglioside/immunology , Natural Killer T-Cells/immunology , Adult , Antigen Presentation/immunology , Antigens, CD1d/metabolism , B-Lymphocytes/metabolism , Cell Line , Cell Proliferation , Cells, Cultured , Coculture Techniques , Flow Cytometry , G(M3) Ganglioside/metabolism , Humans , Ligands , Lymphocyte Activation/immunology , Natural Killer T-Cells/metabolism , Palatine Tonsil/cytology , Protein Binding/immunology
7.
Cytokine ; 77: 14-25, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26517154

ABSTRACT

BACKGROUND/OBJECTIVES: Cirrhosis associated immune dysfunction has been proposed to switch from a pro-inflammatory phenotype in stable cirrhosis to an immunodeficient one in patients with decompensated cirrhosis and acute-on-chronic liver failure. The aim of the present study was to compare serum cytokine levels between healthy patients, stable cirrhosis, and decompensated cirrhotic patients with and without development of acute-on-chronic liver failure (ACLF); and to explore whether any of the measured cytokines is associated with cirrhosis severity and prognosis in ACLF patients. METHODS: Patients were enrolled from October 2013 to May 2014 in two hospitals located in Buenos Aires. Cirrhotic patients with an acute decompensating event were enrolled accordingly to the development of ACLF defined by the CANONIC study group. There were two control groups: healthy subjects (n=14) and stable cirrhotic patients (n=14). Demographic, clinical and biochemical data were obtained. Seventeen cytokines were measured using Bio-Plex Pro Human Cytokine 17-plex Assay. RESULTS: Of the 49 decompensated cirrhotic patients enrolled, 18 (36.7%) developed ACLF. Leukocyte count, MELD score at admission, Clif-SOFA at admission and day 7 were significantly higher in the ACLF group (p=0.046, p<0.001, p<0.001, p<0.001 respectively) as well as short-term mortality (p<0.001) compared to stable and decompensated cirrhotic patients. In comparison with healthy controls, stable cirrhotic and decompensated cirrhotic patients showed increased levels of pro-inflammatory and anti-inflammatory cytokines: IL-6, IL-7, IL-8, IL-10, IL 12, and TNF-α. Decompensated cirrhotic patients with the development of ACLF showed a significant decrease of IL-7, IL-10, IL-12, TNF-α, MCP-1 and IFN-γ, but a sustained response of IL-6 and IL-8. When evaluating cirrhosis severity, IL-6 and IL-8 correlated positively with MELD score, whereas only IL-6 correlated positively with Clif-SOFA score at day 7; IL-2 correlated negatively with Clif-SOFA at admission. In comparison with all scores, leukocyte count showed positive correlation and IFN-γ negative correlation with disease severity. When evaluating survival, only MELD and Clif-SOFA scores had a significant association with mortality. CONCLUSIONS: Pro-inflammatory cytokines and chemo-attractant elements are increased in cirrhosis in comparison with healthy subjects, and display higher values concomitantly with cirrhosis progression. However, in acute-on-chronic liver failure an opposite cytokine pattern that can be resumed as a combination of immune paresis and excessive inflammatory response was observed. Several pro-inflammatory cytokines (IL-2, IL-6, IL-8 and IFN-γ) showed correlation with disease severity; their utility as prognostic biomarkers needs to be further studied.


Subject(s)
Cytokines/blood , Liver Cirrhosis/blood , Liver Cirrhosis/pathology , Severity of Illness Index , Acute-On-Chronic Liver Failure/blood , Acute-On-Chronic Liver Failure/diagnosis , Acute-On-Chronic Liver Failure/mortality , Adult , Biomarkers/blood , Female , Humans , Inflammation Mediators/blood , Liver Cirrhosis/immunology , Male , Middle Aged , Prognosis , Survival Analysis , Survival Rate
8.
J Immunol ; 193(9): 4469-76, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25261474

ABSTRACT

The lack of responsiveness to self and non-self Ags is normally maintained by multiple mechanisms, including the suppressive activities of several T cell subsets. In this study, we show that CD8(+) T cells from both adult peripheral blood and umbilical cord blood mononuclear cells constitutively expressing HLA-DR represent a natural human CD8(+) regulatory T cell subset. Their suppressive effect appears to be cell-to-cell contact dependent and may involve CTLA-4 signaling between neighboring T cells. These regulatory T cells can be expanded in vitro and exhibit a suppressive capacity similar to that observed in ex vivo CD8(+)HLA-DR(+) T cells. The high frequency of CD8(+)HLA-DR(+) T cells that we detected in patients with non-small cell lung cancer deserves further work to confirm their putative suppressor effect within the tumor.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , HLA-DR Antigens/metabolism , Phenotype , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism , Aged , Aged, 80 and over , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/immunology , CTLA-4 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Communication , Cell Proliferation/drug effects , Female , Fetal Blood/cytology , HLA-DR Antigens/immunology , Humans , Immunomodulation , Immunophenotyping , Infant, Newborn , Interleukin-2/pharmacology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation/immunology , Lymphocyte Count , Male , Middle Aged , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology
9.
Pediatr Blood Cancer ; 62(12): 2120-4, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26154941

ABSTRACT

BACKGROUND: Pediatric neuroectodermal malignancies express N-glycolylated gangliosides including N-glycolyl GM3 (NeuGcGM3) as targets for immunotherapy. PROCEDURE: We evaluated the toxicity and maximum tolerated dose and immunological response of racotumomab, an anti-idiotype vaccine targeting NeuGcGM3 through a Phase I study enrolling children with relapsed or resistant tumors expressing NeuGcGM3. MATERIALS AND METHODS: Drug dose was escalated to three levels (0.15-0.25-0.4 mg) of racotumomab administered intradermally. Each drug level included three patients receiving a total of three doses, every 14 days. A confirmation cohort was added to the highest dose level. Antibody response was assessed upon study entry and at 4-week intervals for at least three immunological determinations for each patient. RESULTS: Fourteen patients were enrolled (10 with neuroblastoma, one with retinoblastoma, one with Wilms' tumor, and two with brainstem glioma). Three patients completed the three drug levels and three were enrolled in the confirmation cohort. One patient died of tumor progression before completing the three applications. Racotumomab was well tolerated. The only side effect observed was grade 1-2 toxicity at the injection site. Racotumomab elicited an IgM and/or IgG antibody response directed against NGcGM3 in nine patients and IgM against racotumomab in 11 of 13 evaluable patients. The maximum tolerated dose was not reached and no dose-limiting toxicity was seen. CONCLUSIONS: Racotumomab vaccination has a favorable toxicity profile up to a dose of 0.4 mg, and most patients elicited an immune response. Its activity as immunotherapy for neuroectodermal malignancies will be tested in further clinical trials.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Brain Stem Neoplasms/drug therapy , Cancer Vaccines/administration & dosage , Drug Resistance, Neoplasm/drug effects , Glioma/drug therapy , Neuroblastoma/diet therapy , Wilms Tumor/drug therapy , Antibodies, Monoclonal, Murine-Derived , Antibodies, Neoplasm/blood , Brain Stem Neoplasms/blood , Child , Child, Preschool , Female , Gangliosides/biosynthesis , Gene Expression Regulation, Neoplastic , Glioma/blood , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Infant , Male , Neuroblastoma/blood , Vaccination , Wilms Tumor/blood
10.
Immunol Cell Biol ; 92(10): 882-7, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25047642

ABSTRACT

Human B-cell studies in vitro have routinely used B lymphocytes purified from spleen, blood or tonsils irrespective of potential differences in their immunological traits. In this study, we compared the functional responses of total (CD19(+)) and memory B cells (Bmem; CD19(+)/CD27(+)) isolated from blood and tonsils to different stimuli. Peripheral B cells showed enhanced survival and proliferation compared with their tonsillar equivalents when stimulated for 10 days. Stimulated B cells from both tissues secreted significantly greater amounts of cytokines than unstimulated controls demonstrating their functional responsiveness. Analysis of CD27 expression over time indicated that the conditions that promoted survival and proliferation of peripheral Bmem, caused massive tonsillar Bmem death. Purified tonsillar Bmem failed to expand but rapidly differentiated in antibody secreting cells and subsequently underwent apoptosis. In contrast, circulating Bmem showed delayed activation and differentiation, but exhibited a longer lifespan and active proliferation. In addition, short-term stimulation of tonsillar Bmem resulted in the production of more immunoglobulin G (IgG) than their peripheral counterparts. At later time points, however, IgG production from the different B cells was reversed. Our findings imply that the tissue located and peripheral Bmem have distinct behaviors, indicating organ dependent functional responses that should not be generalizable to all Bmem. This work provides a greater understanding of how Bmem location is coupled to specialized roles of B lymphocytes.


Subject(s)
B-Lymphocyte Subsets/immunology , Immunologic Memory , Palatine Tonsil/immunology , Adult , Apoptosis , Cell Differentiation , Cells, Cultured , Humans , Immunoglobulin G/biosynthesis , Lymphocyte Activation , Palatine Tonsil/cytology
11.
Clin Immunol ; 149(3): 400-10, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24211714

ABSTRACT

It is assumed that the ratio between effector T cells (Teff) and regulatory T cells (Tregs) controls the immune reactivity within the T-cell compartment. The purpose of this study was to investigate if Dexamethasone (Dex) affects Teff and Tregs subsets. Dex induced on Tregs a dose and time-dependent apoptosis which resulted in a relative increase of Teff. After TCR activation, Dex induced a strong proliferative inhibition of Teff, but a weaker proliferative inhibition on Tregs. These effects were modulated by IL-2, which not only restored the proliferative response, but also prevented Dex-induced apoptosis. The highest dose of IL-2 prevented apoptosis on all FOXP3+CD4+ T cells. Meanwhile, the lowest dose only rescued activated Tregs (aTregs), probably related to their CD25 higher expression. Because Dex did not affect the suppressor capacity of aTregs either, our results support the notion that under Dex treatment, the regulatory T-cell compartment maintains its homeostasis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Dexamethasone/pharmacology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Regulatory/drug effects , Adult , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/immunology , Female , Forkhead Transcription Factors/metabolism , Gene Expression , Homeostasis , Humans , Interleukin-2/pharmacology , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Activation/drug effects , Male , Organ Specificity , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology
12.
Glycobiology ; 22(10): 1374-86, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22752006

ABSTRACT

Mechanisms accounting for the protection of the fetal semi-allograft from maternal immune cells remain incompletely understood. In previous studies, we showed that galectin-1 (Gal1), an immunoregulatory glycan-binding protein, hierarchically triggers a cascade of tolerogenic events at the mouse fetomaternal interface. Here, we show that Gal1 confers immune privilege to human trophoblast cells through the modulation of a number of regulatory mechanisms. Gal1 was mainly expressed in invasive extravillous trophoblast cells of human first trimester and term placenta in direct contact with maternal tissue. Expression of Gal1 by the human trophoblast cell line JEG-3 was primarily controlled by progesterone and pro-inflammatory cytokines and impaired T-cell responses by limiting T cell viability, suppressing the secretion of Th1-type cytokines and favoring the expansion of CD4(+)CD25(+)FoxP3(+) regulatory T (T(reg)) cells. Targeted inhibition of Gal1 expression through antibody (Ab)-mediated blockade, addition of the specific disaccharide lactose or retroviral-mediated siRNA strategies prevented these immunoregulatory effects. Consistent with a homeostatic role of endogenous Gal1, patients with recurrent pregnancy loss showed considerably lower levels of circulating Gal1 and had higher frequency of anti-Gal1 auto-Abs in their sera compared with fertile women. Thus, endogenous Gal1 confers immune privilege to human trophoblast cells by triggering a broad tolerogenic program with potential implications in threatened pregnancies.


Subject(s)
Abortion, Habitual/immunology , Galectin 1/immunology , Trophoblasts/immunology , Cell Line , Cell Survival/immunology , Cytokines/immunology , Galectin 1/antagonists & inhibitors , Galectin 1/biosynthesis , Humans , Progesterone/pharmacology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Trophoblasts/cytology
13.
Cancer Cell ; 5(3): 241-51, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15050916

ABSTRACT

Despite the existence of tumor-specific immune cells, most tumors have devised strategies to avoid immune attack. We demonstrate here that galectin-1 (Gal-1), a negative regulator of T cell activation and survival, plays a pivotal role in promoting escape from T cell-dependent immunity, thus conferring immune privilege to tumor cells. Blockade of immunosuppressive Gal-1 in vivo promotes tumor rejection and stimulates the generation of a tumor-specific T cell-mediated response in syngeneic mice, which are then able to resist subsequent challenge with wild-type Gal-1-sufficient tumors. Our data indicate that Gal-1 signaling in activated T cells constitutes an important mechanism of tumor-immune escape and that blockade of this inhibitory signal can allow for and potentiate effective immune responses against tumor cells, with profound implications for cancer immunotherapy.


Subject(s)
Galectin 1/metabolism , Gene Expression Regulation, Neoplastic/physiology , T-Lymphocytes, Cytotoxic/metabolism , Animals , CD4 Antigens/immunology , CD4 Antigens/metabolism , CD8 Antigens/immunology , CD8 Antigens/metabolism , Cell Survival , Galectin 1/immunology , Humans , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice , Microscopy, Fluorescence , Signal Transduction , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured
14.
Am J Reprod Immunol ; 86(2): e13422, 2021 08.
Article in English | MEDLINE | ID: mdl-33730440

ABSTRACT

PROBLEM: Analyze the effect of paternal immunotherapy treatment (PIT) in primary and secondary unexplained recurrent spontaneous abortion (URSA) and unexplained infertility (UI). METHODS OF STUDY: A retrospective study analyzed a two-year follow-up between the generation of MLR-Bfs after PIT treatment (or controls first consultation) and a live birth. Recruited patients included primary URSA with two or more miscarriages at <12 weeks gestation, secondary URSA with previous live birth before two or more miscarriages, and UI with inability to conceive after 2 years of regular unprotected intercourse or in vitro fertilizations (IVF). PIT treated were compared with untreated controls. RESULTS: Primary URSA: live birth was 241/416 (58%) versus 64/282 (23%) controls (p < .0001). Up to age 35, success was 158/217 (73%) and 37/144 (26%) controls (p < .0001). With 3 or more previous URSA, success was 90/135 (67%) versus 17/79 (22%) controls (p < .0001). Between ages 36 and 40, success was 69/147(47%) versus 22/98 (22%) controls (p < .0003), with 3 or more previous URSA live birth was 45/95 (47%) versus 6/46 (13%) controls (p < .0001). In UI, live birth was 99/298 (33%) versus 54/263 (21%) in controls (p < .0009) that increased under age 35 to 53/116 (46%) in treated versus 26/101 (26%) controls (p < .0056). In PIT treated, IVF success required a median of 1 (1.37 ± 0.67) versus a median of 3 IVF procedures (2.75 ± 0.84) in controls. CONCLUSION: PIT is a successful treatment for primary and secondary URSA, and UI. PIT reduced the number of IVF required for achieving pregnancy.


Subject(s)
Abortion, Habitual , Antigens, Neoplasm/blood , Immunotherapy , Infertility, Female , Live Birth , Lymphocyte Transfusion , Abortion, Habitual/blood , Abortion, Habitual/therapy , Adult , Biomarkers/blood , Female , Follow-Up Studies , Humans , Infertility, Female/blood , Infertility, Female/therapy , Lymphocyte Culture Test, Mixed , Retrospective Studies
15.
Clin Immunol ; 136(3): 432-41, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20542739

ABSTRACT

We have previously shown a decreased frequency and function of Tregs in women suffering from recurrent spontaneous abortions (RSA). In the current study, we first investigated the expression of FOXP3 after T-cell activation. We observed that expression of FOXP3 in activated PBMCs was already present above baseline before any cell division, indicating that it was induced in cells that were previously negative for this transcription factor. Because RSA women showed a more limited expansion of FOXP3-positive cells, we next assessed the role of IL-2 signaling through STAT5, which is known to be required for generation of inducible Tregs (iTregs). We demonstrated not only that TGF-beta and IL-2 were diminished but also that the IL-2-STAT-5 signaling axis was down regulated in RSA women. Finally, in addition to a limited FOXP3(+) cells expansion in vitro, iTregs from RSA women showed a strikingly lower suppressor activity.


Subject(s)
Abortion, Habitual/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Case-Control Studies , Female , Follicular Phase/immunology , Forkhead Transcription Factors/metabolism , Humans , Immune Tolerance , In Vitro Techniques , Interleukin-2/metabolism , Interleukin-6/metabolism , Isoantigens/administration & dosage , Kinetics , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , Male , Middle Aged , Pregnancy , STAT5 Transcription Factor/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Young Adult
16.
Clin Immunol ; 134(3): 305-12, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19962351

ABSTRACT

We previously reported that paediatric (PAH) and adult (AAH) forms of type I autoimmune hepatitis (AH) have different HLA-associations and clinical outcome. In the present study we investigated the role of TGF-beta1 genetic polymorphisms in the different outcome of PAH and AAH. We found a significant increase of "high producer" 25GG genotype in PAH and 10CC in AAH. Low inflammation and low fibrosis in AAH was associated with the increase of codon 10CC (high producer) and codon 25CC (low producer) genotypes. The analysis in AAH of the two positions-haplotypes revealed that combined presence of 25GG and 10CC seems to neutralize the 10CC effect which remained in AAH having the 10CC(+)-25GG(-) haplotype. Altogether these results may explain, at least partially, the different clinical outcome of AAH and PAH.


Subject(s)
Hepatitis, Autoimmune/genetics , Hepatitis, Autoimmune/immunology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Alleles , Child , Child, Preschool , Codon , DNA/blood , DNA/genetics , Female , Genotype , Humans , Infant , Male , Middle Aged , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Young Adult
17.
Clin Dev Immunol ; 2010: 814397, 2010.
Article in English | MEDLINE | ID: mdl-21048926

ABSTRACT

Active specific immunotherapy is a promising field in cancer research. N-glycolyl (NGc) gangliosides, and particularly NGcGM3, have received attention as a privileged target for cancer therapy. Many clinical trials have been performed with the anti-NGc-containing gangliosides anti-idiotype monoclonal antibody racotumomab (formerly known as 1E10) and the conjugated NGcGM3/VSSP vaccine for immunotherapy of melanoma, breast, and lung cancer. The present paper examines the role of NGc-gangliosides in tumor biology as well as the available preclinical and clinical data on these vaccine products. A brief discussion on the relevance of prioritization of cancer antigens in vaccine development is also included.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines , G(M3) Ganglioside/analogs & derivatives , Immunotherapy , Neoplasms/immunology , Animals , Antibodies, Anti-Idiotypic/therapeutic use , Antibodies, Monoclonal/therapeutic use , Clinical Trials as Topic , G(M3) Ganglioside/immunology , Humans , Neoplasms/therapy
18.
Cytokine Growth Factor Rev ; 18(1-2): 143-57, 2007.
Article in English | MEDLINE | ID: mdl-17324606

ABSTRACT

From an immunological point of view, the healthy liver has been usually associated with the phenomenon of tolerance. A microenvironment of regulatory cytokines produced by liver Kuppfer cells and liver sinusoidal endothelial cells has contributed, together with resident dendritic cells, to generate a tolerogenic environment in this tissue. In this review we discussed the intrahepatic responses to different sorts of liver injury, such as hepatotrophic viruses, alcohol or putative self-antigens. In each case we analyzed the impact of different cytokines in the clinical outcome of the different pathological situations.


Subject(s)
Cytokines/immunology , Dendritic Cells/immunology , Endothelial Cells/immunology , Immune Tolerance , Kupffer Cells/immunology , Liver Diseases/immunology , Animals , Autoantigens/immunology , Chronic Disease , Dendritic Cells/pathology , Endothelial Cells/pathology , Hepatovirus/immunology , Humans , Kupffer Cells/pathology , Liver/immunology , Liver/injuries , Liver/pathology , Liver Diseases/pathology , Organ Specificity/immunology
20.
Front Immunol ; 9: 2788, 2018.
Article in English | MEDLINE | ID: mdl-30555473

ABSTRACT

We have previously identified a human CD8+HLA-DR+ regulatory T cell subset with the ability to suppress proliferation of autologous PBMCs responder cells through cell contact and CTLA-4 co-inhibitory molecule. The present study characterizes the complete phenotype of CD8+HLA-DR+ Treg cells which showed great similarities with classical CD4+ cells expressing forkhead box P3 (FOXP3). The shared features included the expression of programmed cell death protein 1 (PD-1), T-cell immunoreceptor with Ig and ITIM domains (TIGIT), C-C chemokine receptor type 4 and 5 (CCR4 and CCR5), low expression of CD127, and a memory and effector-like phenotype. CD8+HLA-DR+ Treg-induced suppression on CD8+ responder T cells was abrogated by an anti-PD1 neutralizing antibody. Anti-PD-1 did not abrogate the suppressor effect induced on responder CD4+ T cells. In addition, CD8+HLA-DR+ Treg induced a preferential death on responder CD8+ T cells. This effect was not reversed by PD-1 neutralization. After activation, most CD8+HLA-DR+ Treg acquire programmed death-ligand 1 (PD-L1) expression. Interestingly, PD-L1 may induce apoptosis through CD80 expressed on activated CD8+ responder T cells. After PBMCs stimulation, CD8+HLA-DR+ Treg cells showed an increased frequency of IFN-γ and TNFα positive cells and higher degranulation. These data strongly argue against CD8+HLA-DR+ Treg being exhausted cells. Overall, the data presented in this study indicate that CD8+HLA-DR+ Treg and CD4+FOXP3+ Treg share phenotypic and functional features, which may provide cues to similar involvements in the control of antitumor immune responses and autoimmunity.


Subject(s)
Apoptosis/immunology , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes, Regulatory/immunology , CD8-Positive T-Lymphocytes/cytology , Female , HLA-DR Antigens/immunology , Humans , Interferon-gamma/immunology , Male , T-Lymphocytes, Regulatory/cytology , Tumor Necrosis Factor-alpha/immunology
SELECTION OF CITATIONS
SEARCH DETAIL