Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 141(5): 490-502, 2023 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-36322928

RESUMEN

Primary myelofibrosis (PMF) is a myeloproliferative neoplasm characterized by the clonal expansion of myeloid cells, notably megakaryocytes (MKs), and an aberrant cytokine production leading to bone marrow (BM) fibrosis and insufficiency. Current treatment options are limited. TGF-ß1, a profibrotic and immunosuppressive cytokine, is involved in PMF pathogenesis. While all cell types secrete inactive, latent TGF-ß1, only a few activate the cytokine via cell type-specific mechanisms. The cellular source of the active TGF-ß1 implicated in PMF is not known. Transmembrane protein GARP binds and activates latent TGF-ß1 on the surface of regulatory T lymphocytes (Tregs) and MKs or platelets. Here, we found an increased expression of GARP in the BM and spleen of mice with PMF and tested the therapeutic potential of a monoclonal antibody (mAb) that blocks TGF-ß1 activation by GARP-expressing cells. GARP:TGF-ß1 blockade reduced not only fibrosis but also the clonal expansion of transformed cells. Using mice carrying a genetic deletion of Garp in either Tregs or MKs, we found that the therapeutic effects of GARP:TGF-ß1 blockade in PMF imply targeting GARP on Tregs. These therapeutic effects, accompanied by increased IFN-γ signals in the spleen, were lost upon CD8 T-cell depletion. Our results suggest that the selective blockade of TGF-ß1 activation by GARP-expressing Tregs increases a CD8 T-cell-mediated immune reaction that limits transformed cell expansion, providing a novel approach that could be tested to treat patients with myeloproliferative neoplasms.


Asunto(s)
Mielofibrosis Primaria , Factor de Crecimiento Transformador beta1 , Ratones , Animales , Mielofibrosis Primaria/tratamiento farmacológico , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/metabolismo , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales/metabolismo , Citocinas/metabolismo , Fibrosis , Linfocitos T Reguladores
2.
Cancer Immunol Immunother ; 71(8): 1851-1862, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34973084

RESUMEN

Transmembrane protein GARP binds latent TGF-ß1 to form GARP:(latent)TGF-ß1 complexes on the surface of several cell types including Tregs, B-cells, and platelets. Upon stimulation, these cells release active TGF-ß1. Blocking TGF-ß1 activation by Tregs with anti-GARP:TGF-ß1 mAbs overcomes resistance to PD1/PD-L1 blockade and induces immune-mediated regressions of murine tumors, indicating that Treg-derived TGF-ß1 inhibits anti-tumor immunity. TGF-ß1 exerts a vast array of effects on immune responses. For example, it favors differentiation of TH17 cells and B-cell switch to IgA production, two important processes for mucosal immunity. Here, we sought to determine whether treatment with anti-GARP:TGF-ß1 mAbs would perturb immune responses to intestinal bacterial infection. We observed no aggravation of intestinal disease, no systemic dissemination, and no alteration of innate or adaptative immune responses upon oral gavage of C. rodentium in highly susceptible Il22r-/- mice treated with anti-GARP:TGF-ß1 mAbs. To examine the effects of GARP:TGF-ß1 blockade on Ig production, we compared B cell- and TH cell- responses to OVA or CTB protein immunization in mice carrying deletions of Garp in Tregs, B cells, or platelets. No alteration of adaptive immune responses to protein immunization was observed in the absence of GARP on any of these cells. Altogether, we show that antibody-mediated blockade of GARP:TGF-ß1 or genetic deletion of Garp in Tregs, B cells or platelets, do not alter innate or adaptive immune responses to intestinal bacterial infection or protein immunization in mice. Anti-GARP:TGF-ß1 mAbs, currently tested for cancer immunotherapy, may thus restore anti-tumor immunity without severely impairing other immune defenses. PRéCIS: Immunotherapy with GARP:TGF-ß1 mAbs may restore anti-tumor immunity without impairing immune or inflammatory responses required to maintain homeostasis or host defense against infection, notably at mucosal barriers.


Asunto(s)
Inmunidad Adaptativa , Infecciones Bacterianas , Proteínas de la Membrana , Factor de Crecimiento Transformador beta1 , Animales , Anticuerpos Monoclonales/metabolismo , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/metabolismo , Inmunidad , Inmunización , Proteínas de la Membrana/metabolismo , Ratones , Linfocitos T Reguladores , Factor de Crecimiento Transformador beta1/metabolismo
3.
Environ Microbiol ; 22(6): 1997-2000, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32342578

RESUMEN

The current SARS-CoV-2 pandemic is wreaking havoc throughout the world and has rapidly become a global health emergency. A central question concerning COVID-19 is why some individuals become sick and others not. Many have pointed already at variation in risk factors between individuals. However, the variable outcome of SARS-CoV-2 infections may, at least in part, be due also to differences between the viral subspecies with which individuals are infected. A more pertinent question is how we are to overcome the current pandemic. A vaccine against SARS-CoV-2 would offer significant relief, although vaccine developers have warned that design, testing and production of vaccines may take a year if not longer. Vaccines are based on a handful of different designs (i), but the earliest vaccines were based on the live, attenuated virus. As has been the case for other viruses during earlier pandemics, SARS-CoV-2 will mutate and may naturally attenuate over time (ii). What makes the current pandemic unique is that, thanks to state-of-the-art nucleic acid sequencing technologies, we can follow in detail how SARS-CoV-2 evolves while it spreads. We argue that knowledge of naturally emerging attenuated SARS-CoV-2 variants across the globe should be of key interest in our fight against the pandemic.


Asunto(s)
Betacoronavirus , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo , COVID-19 , Infecciones por Coronavirus , Brotes de Enfermedades , Humanos , Pandemias , Neumonía Viral , SARS-CoV-2
4.
Proc Natl Acad Sci U S A ; 114(47): E10161-E10168, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29109269

RESUMEN

Human regulatory T cells (Tregs) suppress other T cells by converting the latent, inactive form of TGF-ß1 into active TGF-ß1. In Tregs, TGF-ß1 activation requires GARP, a transmembrane protein that binds and presents latent TGF-ß1 on the surface of Tregs stimulated through their T cell receptor. However, GARP is not sufficient because transduction of GARP in non-Treg T cells does not induce active TGF-ß1 production. RGD-binding integrins were shown to activate TGF-ß1 in several non-T cell types. Here we show that αVß8 dimers are present on stimulated human Tregs but not in other T cells, and that antibodies against αV or ß8 subunits block TGF-ß1 activation in vitro. We also show that αV and ß8 interact with GARP/latent TGF-ß1 complexes in human Tregs. Finally, a blocking antibody against ß8 inhibited immunosuppression by human Tregs in a model of xenogeneic graft-vs.-host disease induced by the transfer of human T cells in immunodeficient mice. These results show that TGF-ß1 activation on the surface of human Tregs implies an interaction between the integrin αVß8 and GARP/latent TGF-ß1 complexes. Immunosuppression by human Tregs can be inhibited by antibodies against GARP or against the integrin ß8 subunit. Such antibodies may prove beneficial against cancer or chronic infections.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Tolerancia Inmunológica/efectos de los fármacos , Integrinas/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta1/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Integrinas/antagonistas & inhibidores , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones SCID , Neoplasias/inmunología , Neoplasias/terapia , Linfocitos T Reguladores/trasplante , Factor de Crecimiento Transformador beta1/metabolismo , Trasplante Heterólogo
5.
J Immunol ; 199(2): 391-396, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28607112

RESUMEN

Production of active TGF-ß is regulated at a posttranslational level and implies release of the mature cytokine dimer from the inactive, latent TGF-ß precursor. There are several cell-type specific mechanisms of TGF-ß activation. We identified a new mechanism operating on the surface of human regulatory T cells and involving membrane protein GARP, which binds latent TGF-ß1. The paracrine activity of regulatory T cell-derived TGF-ß1 contributes to immunosuppression and can be inhibited with anti-GARP Abs. Whether other immune cell types use surface GARP to activate latent TGF-ß1 was not known. We show in this study that stimulated, human B lymphocytes produce active TGF-ß1 from surface GARP/latent TGF-ß1 complexes with isotype switching to IgA production.


Asunto(s)
Linfocitos B/inmunología , Inmunoglobulina A/biosíntesis , Cambio de Clase de Inmunoglobulina , Proteínas de la Membrana/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Linfocitos B/efectos de los fármacos , Citocinas/inmunología , Humanos , Inmunoglobulina A/inmunología , Activación de Linfocitos , Oligodesoxirribonucleótidos/inmunología , Unión Proteica , Recombinación Genética , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta1/inmunología
6.
Mol Hum Reprod ; 24(12): 602-612, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30371828

RESUMEN

STUDY QUESTION: Do all 10 human pregnancy-specific beta 1-glycoproteins (PSGs) and murine PSG23 activate latent transforming growth factor-ß1 (TGF-ß1)? SUMMARY ANSWER: All human PSGs and murine PSG23 activated latent TGF-ß1. WHAT IS KNOWN ALREADY: Two of the 10 members of the PSG1 family, PSG1 and PSG9, were previously shown to activate the soluble small latent complex of TGF-ß1, a cytokine with potent immune suppressive functions. STUDY DESIGN, SIZE, DURATION: Recombinant PSGs were generated and tested for their ability to activate the small latent complex of TGF-ß1 in a cell-free ELISA-based assay and in a bioassay. In addition, we tested the ability of PSG1 and PSG4 to activate latent TGF-ß bound to the extracellular matrix (ECM) or on the membranes of the Jurkat human T-cell line. PARTICIPANTS/MATERIALS, SETTING, METHODS: Recombinant PSGs were generated by transient transfection and purified with a His-Trap column followed by gel filtration chromatography. The purified PSGs were compared to vehicle (PBS) used as control for their ability to activate the small latent complex of TGF-ß1. The concentration of active TGF-ß was measured in an ELISA using the TGF-ß receptor II as capture and a bioassay using transformed mink epithelial cells that express luciferase in response to active TGF-ß. The specificity of the signal was confirmed using a TGF-ß receptor inhibitor. We also measured the binding kinetics of some human PSGs for the latent-associated peptide (LAP) of TGF-ß using surface plasmon resonance and determined whether PSG1 and PSG4 could activate the large latent complex of TGF-ß1 bound to the ECM and latent TGF-ß1 bound to the cell membrane. All experiments were performed in triplicate wells and repeated three times. MAIN RESULTS AND THE ROLE OF CHANCE: All human PSGs activated the small latent complex of TGF-ß1 (P < 0.05 vs. control) and showed similar affinities (KD) for LAP. Despite the lack of sequence conservation with its human counterparts, the ability to activate latent TGF-ß1 was shared by a member of the murine PSG family. We found that PSG1 and PSG4 activated the latent TGF-ß stored in the ECM (P < 0.01) but did not activate latent TGF-ß1 bound to glycoprotein A repetitions predominant (GARP) on the surface of Jurkat T cells. LIMITATIONS, REASONS FOR CAUTION: The affinity of the interaction of LAP and PSGs was calculated using recombinant proteins, which may differ from the native proteins in their post-translational modifications. We also utilized a truncated form of murine PSG23 rather than the full-length protein. For the studies testing the ability of PSGs to activate membrane-bound TGF-ß1, we utilized the T-cell line Jurkat and Jurkat cells expressing GARP rather than primary T regulatory cells. All the studies were performed in vitro. WIDER IMPLICATIONS OF THE FINDINGS: Here, we show that all human PSGs activate TGF-ß1 and that this function is conserved in at least one member of the rodent PSG family. In vivo PSGs could potentially increase the availability of active TGF-ß1 from the soluble and matrix-bound latent forms of the cytokine contributing to the establishment of a tolerogenic environment during pregnancy. LARGE-SCALE DATA: None. STUDY FUNDING/COMPETING INTEREST(S): The research was supported by a grant from the Collaborative Health Initiative Research Program (CHIRP). No conflicts of interests are declared by the authors.


Asunto(s)
Glicoproteínas beta 1 Específicas del Embarazo/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Ensayo de Inmunoadsorción Enzimática , Matriz Extracelular/metabolismo , Femenino , Heparitina Sulfato , Humanos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Embarazo , Glicoproteínas beta 1 Específicas del Embarazo/genética , Factor de Crecimiento Transformador beta1/genética
7.
J Immunol ; 195(3): 832-40, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26071562

RESUMEN

Follicular regulatory T cells (TFR) have been extensively characterized in mice and participate in germinal center responses by regulating the maturation of B cells and production of (auto)antibodies. We report that circulating TFR are phenotypically distinct from tonsil-derived TFR in humans. They have a lower expression of follicular markers, and display a memory phenotype and lack of high expression of B cell lymphoma 6 and ICOS. However, the suppressive function, expression of regulatory markers, and FOXP3 methylation status of blood TFR is comparable with tonsil-derived TFR. Moreover, we show that circulating TFR frequencies increase after influenza vaccination and correlate with anti-flu Ab responses, indicating a fully functional population. Multiple sclerosis (MS) was used as a model for autoimmune disease to investigate alterations in circulating TFR. MS patients had a significantly lower frequency of circulating TFR compared with healthy control subjects. Furthermore, the circulating TFR compartment of MS patients displayed an increased proportion of Th17-like TFR. Finally, TFR of MS patients had a strongly reduced suppressive function compared with healthy control subjects. We conclude that circulating TFR are a circulating memory population derived from lymphoid resident TFR, making them a valid alternative to investigate alterations in germinal center responses in the context of autoimmune diseases, and TFR impairment is prominent in MS.


Asunto(s)
Antígeno de Maduración de Linfocitos B/biosíntesis , Vacunas contra la Influenza/inmunología , Esclerosis Múltiple/inmunología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Adulto , Anticuerpos/sangre , Linfocitos B/inmunología , Femenino , Factores de Transcripción Forkhead/metabolismo , Humanos , Memoria Inmunológica/inmunología , Proteína Coestimuladora de Linfocitos T Inducibles/biosíntesis , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Masculino , Metilación , Esclerosis Múltiple/sangre , Vacunación , Adulto Joven
8.
J Biol Chem ; 290(33): 20105-16, 2015 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-26126825

RESUMEN

Production of active TGF-ß1 is one mechanism by which human regulatory T cells (Tregs) suppress immune responses. This production is regulated by glycoprotein A repetitions predominant (GARP), a transmembrane protein present on stimulated Tregs but not on other T lymphocytes (Th and CTLs). GARP forms disulfide bonds with proTGF-ß1, favors its cleavage into latent inactive TGF-ß1, induces the secretion and surface presentation of GARP·latent TGF-ß1 complexes, and is required for activation of the cytokine in Tregs. We explored whether additional Treg-specific protein(s) associated with GARP·TGF-ß1 complexes regulate TGF-ß1 production in Tregs. We searched for such proteins by yeast two-hybrid assay, using GARP as a bait to screen a human Treg cDNA library. We identified lysosomal-associated transmembrane protein 4B (LAPTM4B), which interacts with GARP in mammalian cells and is expressed at higher levels in Tregs than in Th cells. LAPTM4B decreases cleavage of proTGF-ß1, secretion of soluble latent TGF-ß1, and surface presentation of GARP·TGF-ß1 complexes by Tregs but does not contribute to TGF-ß1 activation. Therefore, LAPTM4B binds to GARP and is a negative regulator of TGF-ß1 production in human Tregs. It may play a role in the control of immune responses by decreasing Treg immunosuppression.


Asunto(s)
Proteínas de la Membrana/fisiología , Proteínas Oncogénicas/fisiología , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta1/biosíntesis , Células HEK293 , Humanos , Proteínas de la Membrana/metabolismo , Técnicas del Sistema de Dos Híbridos
9.
Brain Behav Immun ; 45: 180-8, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25514345

RESUMEN

Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS), for which current treatments are unable to prevent disease progression. Based on its neuroprotective and neuroregenerating properties, leukemia inhibitory factor (LIF), a member of the interleukin-6 (IL-6) cytokine family, is proposed as a novel candidate for MS therapy. However, its effect on the autoimmune response remains unclear. In this study, we determined how LIF modulates T cell responses that play a crucial role in the pathogenesis of MS. We demonstrate that expression of the LIF receptor was strongly increased on immune cells of MS patients. LIF treatment potently boosted the number of regulatory T cells (Tregs) in CD4(+) T cells isolated from healthy controls and MS patients with low serum levels of IL-6. Moreover, IL-6 signaling was reduced in the donors that responded to LIF treatment in vitro. Our data together with previous findings revealing that IL-6 inhibits Treg development, suggest an opposing function of LIF and IL-6. In a preclinical animal model of MS we shifted the LIF/IL-6 balance in favor of LIF by CNS-targeted overexpression. This increased the number of Tregs in the CNS during active autoimmune responses and reduced disease symptoms. In conclusion, our data show that LIF downregulates the autoimmune response by enhancing Treg numbers, providing further impetus for the use of LIF as a novel treatment for MS and other autoimmune diseases.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Interleucina-6/inmunología , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/inmunología , Factor Inhibidor de Leucemia/inmunología , Esclerosis Múltiple/inmunología , Linfocitos T Reguladores/inmunología , Adulto , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Estudios de Casos y Controles , Femenino , Humanos , Técnicas In Vitro , Interleucina-6/metabolismo , Factor Inhibidor de Leucemia/farmacología , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Linfocitos T Reguladores/efectos de los fármacos
10.
Transfusion ; 54(2): 353-63, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23772685

RESUMEN

BACKGROUND: We investigated the ability of clinical-grade enriched human regulatory T cells (Treg) to attenuate experimental xenogeneic graft-versus-host disease (GVHD) induced by peripheral blood mononuclear cells (PBMNCs; autologous to Treg) infusion in NSG mice, as well as verified their inability to induce xenogeneic GVHD when infused alone. STUDY DESIGN AND METHODS: Human Treg were isolated from peripheral blood apheresis products with a cell separation system (CliniMACS, Miltenyi Biotec GmbH) using a two-step procedure (simultaneous CD8 and CD19 depletion followed by CD25-positive selection) in six independent experiments with six different healthy volunteer donors. Sublethally (2.5 Gy) irradiated NSG mice were given 2 × 10(6) cytapheresis (PBMNC) product cells intravenously (IV) without (PBMNC group) or with 1 × 10(6) Treg (PBMNC + Treg group), while other NSG mice received 2 × 10(6) enriched Treg alone (also in IV; Treg group). RESULTS: The first five procedures were successful at obtaining a relatively pure Treg population (defined as >50%), while the sixth procedure, due to a technical problem, was not (Treg purity, 42%). Treg cotransfusion significantly delayed death from xenogeneic GVHD in the first five experiments, (p < 0.0001) but not in the sixth experiment. Importantly, none of the mice given enriched Treg alone (Treg group) experienced clinical signs of GVHD, while, interestingly, the CD4+ cells found in these mice 26 days after transplantation were mainly conventional T cells (median CD25+FoxP3+ cells among human CD4+ total cells were only 2.1, 3.1, and 12.2% in spleen, marrow, and blood, respectively). CONCLUSIONS: Infusion of clinical-grade enriched Treg delayed the occurrence of xenogeneic GVHD without inducing toxicity in this murine model.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/prevención & control , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/trasplante , Animales , Eliminación de Componentes Sanguíneos/métodos , Modelos Animales de Enfermedad , Humanos , Inmunofenotipificación , Ratones , Ratones Endogámicos NOD , Ratones SCID , Factores de Tiempo , Trasplante Heterólogo
11.
Personal Ment Health ; 2023 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-37997227

RESUMEN

Borderline personality disorder (BPD) is a severe mental health condition marked by impairments in self and interpersonal functioning. Stigma from health staff may often result in a reluctance to diagnose, impacting recovery trajectories. Qualitative interviews were conducted with participants (N = 15; M Age = 36.4 years, SD = 7.5; 93.3% female) with lived experience of BPD exploring topics of illness onset, insight, experience of diagnosis and treatment. Qualitative responses were analysed within a co-design framework with a member of the research team who identifies as having a lived experience of BPD. On average, participant symptoms emerged at 12.1 years of age (SD = 6.6 years, range 1.5-27), but diagnoses of BPD were delayed until 30.2 years (SD = 7.8 years, range 18-44) resulting in a 'diagnosis gap' of 18.1 years (SD = 9.6 years, range 3-30). Participant explanations for BPD emergence varied from biological, psychological and social factors. Benefits of diagnosis (e.g., fostering insight, aiding treatment planning and reducing isolation) were contrasted with challenges (e.g., stigma and treatment unavailability). Delay in diagnosis was common, and no participants reported receiving a diagnosis of BPD during their adolescence yet 85% felt they would have benefited from a diagnosis in adolescence. Only a quarter (27%) felt highly supported in the diagnostic process. An ideal four-step diagnosis procedure was outlined based on recommendations from participants with a lived experience; this involved the following: (1) explain the process, (2) assess thoroughly, (3) explore how the features are active in everyday life and (4) link diagnosis to evidence-based treatment planning.

12.
Cancers (Basel) ; 15(23)2023 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-38067325

RESUMEN

Several types of cancer spread through the lymphatic system via the sentinel lymph nodes (LNs). Such LN-draining primary tumors, modified by tumor factors, lead to the formation of a metastatic niche associated with an increased number of Foxp3+ regulatory T cells (Tregs). These cells are expected to contribute to the elaboration of an immune-suppressive environment. Activated Tregs express glycoprotein A repetitions predominant (GARP), which binds and presents latent transforming growth factor beta 1 (TGF-ß1) at their surface. GARP is also expressed by other non-immune cell types poorly described in LNs. Here, we mapped GARP expression in non-immune cells in human and mouse metastatic LNs. The mining of available (human and murine) scRNA-Seq datasets revealed GARP expression by blood (BEC)/lymphatic (LEC) endothelial, fibroblastic, and perivascular cells. Consistently, through immunostaining and in situ RNA hybridization approaches, GARP was detected in and around blood and lymphatic vessels, in (αSMA+) fibroblasts, and in perivascular cells associated with an abundant matrix. Strikingly, GARP was detected in LECs forming the subcapsular sinus and high endothelial venules (HEVs), two vascular structures localized at the interface between LNs and the afferent lymphatic and blood vessels. Altogether, we here provide the first distribution maps for GARP in human and murine LNs.

13.
Lupus Sci Med ; 10(2)2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37899089

RESUMEN

OBJECTIVES: Despite treatment, one-third of patients with lupus nephritis (LN) show a decline in renal function. Prognostic markers of poor outcome as well as novel therapeutic targets are therefore highly sought. We showed that p16INK4a, a marker of cellular senescence, is observed in baseline kidney biopsies from patients with LN, and is associated with renal disease. Here, we set out to assess for whether these findings are recapitulated in the B6.NZMSle1/Sle2/Sle3 (B6.Sle1.2.3) mouse model of spontaneous lupus. METHODS: We evaluated the occurrence and time of onset of p16Ink4a staining by immunohistochemistry on kidney sections, and tested for its association with multiple renal and systemic disease parameters, fibrosis and CD8+ T cell infiltration, in two cohorts of B6.Sle1.2.3 mice. RESULTS: The presence of p16Ink4a-positive cells in kidney was significantly associated with increased urine albumin/creatinine ratio, histopathological scores, CD8+ T cell infiltration and fibrosis, in both B6.Sle1.2.3 cohorts. In contrast, p16Ink4a staining was not associated with systemic disease parameters. A time course showed that systemic disease parameters as well as glomerular IgG deposits appeared in B6.Sle1.2.3 mice by 4 months of age; the appearance of p16Ink4a-positive cells occurred later, by 8 months of age, overlapping with renal disease. CONCLUSION: We report, for the first time, the presence of p16Ink4a-positive cells, a marker of cellular senescence, in the B6.Sle1.2.3 kidney, and their association with renal disease severity. This provides a preclinical model in which to test for the role of cellular senescence in the pathogenesis of LN, as a potential kidney-intrinsic disease mechanism.


Asunto(s)
Lupus Eritematoso Sistémico , Nefritis Lúpica , Ratones , Humanos , Animales , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Lupus Eritematoso Sistémico/complicaciones , Lupus Eritematoso Sistémico/patología , Riñón/patología , Nefritis Lúpica/patología , Senescencia Celular , Fibrosis
14.
Int J Cancer ; 130(8): 1960-6, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21618521

RESUMEN

The human suppressive T cells that stably express transcription factor FOXP3, or regulatory T cells (Tregs), are thought to suppress antitumor immune responses. The most specific marker for human Tregs is the demethylation of CpG dinucleotides located in the first intron of FOXP3 (FOXP3i1). FOXP3i1 is completely methylated in other hematopoietic cells, including nonsuppressive T cells that transiently express FOXP3 after activation. Previously, we and others reported estimations of the frequency of Tregs in the blood of melanoma patients using a FOXP3i1 methylation-specific qPCR assay. Here, we attempted to quantify Tregs inside tumor samples using this assay. However, we found demethylated FOXP3i1 sequences in the melanoma cells themselves. This demethylation was not associated with substantial FOXP3 mRNA or protein expression, even though the demethylation extended to the promoter and terminal regions of the gene in some melanoma cells. Our results imply that analyzing Treg frequencies by quantification of demethylated FOXP3i1 will require that tumor-infiltrating T cells be separated from melanoma cells.


Asunto(s)
Metilación de ADN , Factores de Transcripción Forkhead/genética , Melanoma/genética , Linfocitos T Reguladores/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Islas de CpG/genética , Epigénesis Genética , Femenino , Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Intrones/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Melanoma/metabolismo , Melanoma/patología , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Smad2/genética , Proteína Smad2/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Linfocitos T Reguladores/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología
15.
Am J Respir Crit Care Med ; 184(11): 1270-81, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21868503

RESUMEN

RATIONALE: There is evidence that CD4(+) effector T lymphocytes (T eff) participate in the development of lung fibrosis, but the role of their CD4(+) regulatory T-cell (T reg) counterparts remains to be determined. OBJECTIVES: To elucidate the contribution of T reg cells in a mouse model of lung fibrosis induced by silica (SiO(2)) particles. METHODS: Lung T reg and T eff cells purified from SiO(2)-treated Foxp3-GFP transgenic mice were cocultured with naive lung fibroblasts or transferred to the lungs of healthy mice. DEREG mice, which express the diphtheria toxin receptor under the control of the foxp3 gene, were used to deplete T reg cells during fibrogenesis. MEASUREMENTS AND MAIN RESULTS: CD4(+) Foxp3(+) T reg cells were persistently recruited in the lungs in response to SiO(2). T reg accumulation paralleled the establishment of pulmonary immunosuppression and fibrosis. T reg cells highly expressed platelet-derived growth factor (PDGF)-B via a TGF-ß autocrine signaling pathway, directly stimulated fibroblast proliferation in vitro, and increased lung collagen deposition upon transfer in the lung of naive mice. The direct profibrotic effects of T reg cells were abolished by the inhibitor of the PDGF-B/TGF-ß signaling pathway, imatinib mesylate. Neutralization of T reg-immunosuppressive activity resulted in enhanced accumulation of T eff cells and IL-4-driven pulmonary fibrogenesis, further demonstrating that T reg cells control T eff cell functions during inflammatory fibrosis. CONCLUSIONS: Our study indicates that T reg cells contribute to lung fibrosis by stimulating fibroblasts through the secretion of PDGF-B in noninflammatory conditions and regulate detrimental T eff cell activities during inflammation-related fibrosis.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Factores de Transcripción Forkhead/inmunología , Factor de Crecimiento Derivado de Plaquetas/inmunología , Fibrosis Pulmonar/inmunología , Linfocitos T Reguladores/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Técnicas de Cultivo de Célula , Modelos Animales de Enfermedad , Citometría de Flujo/métodos , Factores de Transcripción Forkhead/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Fibrosis Pulmonar/metabolismo , Linfocitos T Reguladores/metabolismo
16.
Cancers (Basel) ; 14(19)2022 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-36230610

RESUMEN

Papillary thyroid carcinoma (PTC) is the most frequent histological subtype of thyroid cancers (TC), and BRAFV600E genetic alteration is found in 60% of this endocrine cancer. This oncogene is associated with poor prognosis, resistance to radioiodine therapy, and tumor progression. Histological follow-up by anatomo-pathologists revealed that two-thirds of surgically-removed thyroids do not present malignant lesions. Thus, continued fundamental research into the molecular mechanisms of TC downstream of BRAFV600E remains central to better understanding the clinical behavior of these tumors. To study PTC, we used a mouse model in which expression of BRAFV600E was specifically switched on in thyrocytes by doxycycline administration. Upon daily intraperitoneal doxycycline injection, thyroid tissue rapidly acquired histological features mimicking human PTC. Transcriptomic analysis revealed major changes in immune signaling pathways upon BRAFV600E induction. Multiplex immunofluorescence confirmed the abundant recruitment of macrophages, among which a population of LYVE-1+/CD206+/STABILIN-1+ was dramatically increased. By genetically inactivating the gene coding for the scavenger receptor STABILIN-1, we showed an increase of CD8+ T cells in this in situ BRAFV600E-dependent TC. Lastly, we demonstrated the presence of CD206+/STABILIN-1+ macrophages in human thyroid pathologies. Altogether, we revealed the recruitment of immunosuppressive STABILIN-1 macrophages in a PTC mouse model and the interest to further study this macrophage subpopulation in human thyroid tissues.

17.
Biochem Pharmacol ; 192: 114697, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34302795

RESUMEN

The TGF-ß1 cytokine is a key mediator of many biological processes. Complex regulatory mechanisms are in place that allow one single molecule to exert so many distinct indispensable activities. The complexity of TGF-ß1 biology is further illustrated by the opposing dual roles it plays during cancer progression. Risks of toxicities combined with lack of convincing therapeutical efficacy explain at least in part why therapies targeting TGF-ß1 have lagged behind in past decades. However, recent successes of immunostimulatory antibodies for the immunotherapy of cancer and findings that TGF-ß1 activity associates with resistance to immunotherapeutic drugs have revived the field. In this review, we discuss the biology of TGF-ß1 with a special focus on its roles in regulating immune responses in the context of cancer. We describe the various therapeutic approaches available to inhibit TGF-ß signalling, and more recent findings that allow selective targeting of specific sources of TGF-ß activity, which may prove relevant to increase the efficacy and reduce the toxicity of cancer immunotherapy.


Asunto(s)
Inmunidad Celular/inmunología , Terapia de Inmunosupresión/métodos , Inmunoterapia/métodos , Neoplasias/inmunología , Factor de Crecimiento Transformador beta1/inmunología , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/inmunología , Humanos , Neoplasias/terapia , Estructura Secundaria de Proteína , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/química
18.
Front Immunol ; 12: 704050, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34386010

RESUMEN

When combined with anti-PD-1, monoclonal antibodies (mAbs) against GARP:TGF-ß1 complexes induced more frequent immune-mediated rejections of CT26 and MC38 murine tumors than anti-PD-1 alone. In both types of tumors, the activity of anti-GARP:TGF-ß1 mAbs resulted from blocking active TGF-ß1 production and immunosuppression by GARP-expressing regulatory T cells. In CT26 tumors, combined GARP:TGF-ß1/PD-1 blockade did not augment the infiltration of T cells, but did increase the effector functions of already present anti-tumor T cells. Here we show that, in contrast, in MC38, combined GARP:TGF-ß1/PD-1 blockade increased infiltration of T cells, as a result of increased extravasation of T cells from blood vessels. Unexpectedly, combined GARP:TGF-ß1/PD-1 blockade also increased the density of GARP+ blood vessels covered by pericytes in MC38, but not in CT26 tumors. This appears to occur because anti-GARP:TGF-ß1, by blocking TGF-ß1 signals, favors the proliferation of and expression of adhesion molecules such as E-selectin by blood endothelial cells. The resulting densification of intratumoral blood vasculature probably contributes to increased T cell infiltration and to the therapeutic efficacy of GARP:TGF-ß1/PD-1 blockade in MC38. We conclude from these distinct observations in MC38 and CT26, that the combined blockades of GARP:TGF-ß1 and PD-1 can exert anti-tumor activity via multiple mechanisms, including the densification and normalization of intratumoral blood vasculature, the increase of T cell infiltration into the tumor and the increase of the effector functions of intratumoral tumor-specific T cells. This may prove important for the selection of cancer patients who could benefit from combined GARP:TGF-ß1/PD-1 blockade in the clinics.


Asunto(s)
Antineoplásicos Inmunológicos , Vasos Sanguíneos/inmunología , Proteínas de la Membrana , Neoplasias Experimentales , Neovascularización Patológica , Pericitos/inmunología , Receptor de Muerte Celular Programada 1 , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta1 , Animales , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/farmacología , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/inmunología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/inmunología
19.
J Immunother Cancer ; 9(2)2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33568351

RESUMEN

BACKGROUND: Natural killer (NK) cells provide important immune protection from cancer and are a key requirement for particular immunotherapies. There is accumulating evidence that NK cells become dysfunctional during cancer. Overcoming NK cell exhaustion would be an important step to allow them to function optimally in a range of NK cell therapies, including those that depend on autologos circulating NK cells. We have previously demonstrated that NK cells undergo a normal metabolic reprogramming in response to cytokine activation and that this is required for optimal function. The objective of this work was to investigate if cellular metabolism of circulating NK cells is dysregulated in patients with metastatic breast cancer and if so, to gain insights into potential mechanisms underpinning this. Such discoveries would provide important insights into how to unleash the full activity of NK cells for maximum immunotherapy output. METHODS: Single-cell analysis, metabolic flux and confocal analysis of NK cells from patients with metastatic breast cancer and healthy controls RESULTS: In addition to reduced interferon-γ production and cytotoxicity, peripheral blood NK cells from patients had clear metabolic deficits including reduced glycolysis and oxidative phosphorylation. There were also distinct morphologically alterations in the mitochondria with increased mitochondrial fragmentation observed. Transforminggrowth factor-ß (TGFß) was identified as a key driver of this phenotype as blocking its activity reversed many metabolic and functional readouts. Expression of glycoprotein-A repetitions predominant (GARP) and latency associated peptide (LAP), which are involved with a novel TGFß processing pathway, was increased on NK cells from some patients. Blocking the GARP-TGFß axis recapitulated the effects of TGFß neutralization, highlighting GARP as a novel NK cell immunotherapy target for the first time. CONCLUSIONS: TGFß contributes to metabolic dysfunction of circulating NK cells in patients with metastatic breast cancer. Blocking TGFß and/or GARP can restore NK cell metabolism and function and is an important target for improving NK cell-based immunotherapies.


Asunto(s)
Neoplasias de la Mama/metabolismo , Metabolismo Energético , Células Asesinas Naturales/metabolismo , Mitocondrias/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Estudios de Casos y Controles , Técnicas de Cocultivo , Citotoxicidad Inmunológica , Femenino , Glucólisis , Humanos , Interferón gamma/metabolismo , Células K562 , Células Asesinas Naturales/inmunología , Proteínas de la Membrana , Microscopía Confocal , Persona de Mediana Edad , Mitocondrias/inmunología , Metástasis de la Neoplasia , Fosforilación Oxidativa , Transducción de Señal , Análisis de la Célula Individual , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo
20.
J Clin Invest ; 131(19)2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34411002

RESUMEN

We previously demonstrated that tumor-infiltrating lymphocytes (TIL) in human breast cancer sometimes form organized tertiary lymphoid structures (TLS) characterized by CXCL13-producing T follicular helper (Tfh) cells. The present study found that CD4+ Tfh TIL, CD8+ TIL, and TIL-B, colocalizing in TLS, all express the CXCL13 receptor CXCR5. An ex vivo functional assay determined that only activated, functional Th1-oriented Tfh TIL (PD-1hiICOSint phenotype) provide help for immunoglobulin and IFN-γ production. A functional Tfh TIL presence signals an active TLS, characterized by humoral (immunoglobulins, Ki-67+ TIL-B in active germinal centers) and cytotoxic (GZMB+CD8+ and GZMB+CD68+ TIL plus Th1 gene expression) immune responses. Analysis of active versus inactive TLS in untreated patients revealed that the former are associated with positive clinical outcomes. TLS also contain functional T follicular regulatory (Tfr) TIL, which are characterized by a CD25+CXCR5+GARP+FOXP3+ phenotype and a demethylated FOXP3 gene. Functional Tfr inhibited functional Tfh activities via a glycoprotein A repetitions predominant (GARP)-associated TGF-ß-dependent mechanism. The activity of tumor-associated TLS was dictated by the relative balance between functional Tfh TIL and functional Tfr TIL. These data provide mechanistic insight into TLS processes orchestrated by functional Th1-oriented Tfh TIL, including TIL-B and CD8+ TIL activation and immunological memory generation. Tfh TIL, regulated by functional Tfr TIL, are an expected key target of PD-1/PD-L1 blockade.


Asunto(s)
Neoplasias de la Mama/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Células T Auxiliares Foliculares/inmunología , Células TH1/inmunología , Inmunidad Adaptativa , Linfocitos T CD8-positivos/inmunología , Femenino , Humanos , Proteínas de la Membrana/análisis , Proteínas de la Membrana/fisiología , Receptor de Muerte Celular Programada 1/análisis , Receptores CXCR5/análisis , Linfocitos T Reguladores/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA