Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Eur J Immunol ; 48(7): 1137-1152, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29624673

RESUMEN

The bone marrow hosts NK cells whose distribution, motility and response to systemic immune challenge are poorly understood. At steady state, two-photon microscopy of the bone marrow in Ncr1gfp/+ mice captured motile NK cells interacting with dendritic cells. NK cells expressed markers and effector molecules of mature cells. Following poly (I:C) injection, RNA-Seq of NK cells revealed three phases of transcription featuring immune response genes followed by posttranscriptional processes and proliferation. Functionally, poly (I:C) promoted upregulation of granzyme B, enhanced cytotoxicity in vitro and in vivo, and, in the same individual cells, triggered proliferation. Two-photon imaging revealed that the proportion of sinusoidal NK cells decreased, while at the same time parenchymal NK cells accelerated, swelled and divided within the bone marrow. MVA viremia induced similar responses. Our findings demonstrate that the bone marrow is patrolled by mature NK cells that rapidly proliferate in response to systemic viral challenge while maintaining their effector functions.


Asunto(s)
Médula Ósea/inmunología , Células Dendríticas/inmunología , Células Asesinas Naturales/inmunología , Viremia/inmunología , Animales , Antígenos Ly/genética , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Citotoxicidad Inmunológica , Granzimas/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptor 1 Gatillante de la Citotoxidad Natural/genética , Poli I-C/inmunología , Activación Viral
2.
Cancer Immunol Immunother ; 68(8): 1287-1301, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31253998

RESUMEN

Patchy infiltration of tumors by cytotoxic T cells (CTLs) predicts poorer prognosis for cancer patients. The factors limiting intratumoral CTL dissemination, though, are poorly understood. To study CTL dissemination in tumors, we histologically examined human melanoma samples and used mice to image B16-OVA tumors infiltrated by OT-I CTLs using intravital two-photon microscopy. In patients, most CTLs concentrated around peripheral blood vessels, especially in poorly infiltrated tumors. In mice, OT-I CTLs had to cluster around tumor cells to efficiently kill them in a contact-and perforin-dependent manner and cytotoxicity was strictly antigen-specific. OT-I CTLs as well as non-specific CTLs concentrated around peripheral vessels, and cleared the tumor cells around them. This was also the case when CTLs were injected directly into the tumors. CTLs crawled rapidly only in areas within 50 µm of flowing blood vessels and transient occlusion of vessels immediately, though reversibly, stopped their migration. In vitro, oxygen depletion and blockade of oxidative phosphorylation also reduced CTL motility. Taken together, these results suggest that hypoxia limits CTL migration away from blood vessels, providing immune-privileged niches for tumor cells to survive. Normalizing intratumoral vasculature may thus synergize with tumor immunotherapy.


Asunto(s)
Vasos Sanguíneos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma/inmunología , Neoplasias Cutáneas/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Antígenos de Neoplasias/inmunología , Movimiento Celular , Citotoxicidad Inmunológica , Humanos , Melanoma/irrigación sanguínea , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales , Neovascularización Patológica , Fosforilación Oxidativa , Perforina/metabolismo , Neoplasias Cutáneas/irrigación sanguínea
3.
Eur J Immunol ; 47(10): 1802-1818, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28872666

RESUMEN

Antigen (Ag) specific activation of naïve T cells by migrating dendritic cells (DCs) is a highly efficient process, although the chances for their colocalization in lymph nodes (LNs) appear low. Ag presentation may be delegated from Ag-donor DCs to the abundant resident DCs, but the routes of Ag transfer and how it facilitates T-cell activation remain unclear. We visualized CD8+ T cell-DC interactions to study the sites, routes, and cells mediating Ag transfer in mice. In vitro, Ag transfer from isolated ovalbumin (OVA)+ bone marrow (BM) DCs triggered widespread arrest, Ca2+ flux, and CD69 upregulation in OT-I T cells contacting recipient DCs. Intravital two-photon imaging revealed that survival of Ag-donor DCs in LNs was required for Ag dissemination among resident CD11c+ DCs. Upon interaction with recipient DCs, CD8+ T cells clustered, upregulated CD69, proliferated and differentiated into effectors. Few DCs sufficed for activation, and for efficient Ag dissemination lymphocyte function associated antigen 1 (LFA-1) expression on recipient DCs was essential. Similar findings characterized DCs infected with a replication-deficient OVA-expressing Vaccinia virus known to downregulate MHC-I. Overall, active Ag dissemination from live incoming DCs helped activate CD8+ T cells by increasing the number of effective presenting cells and salvaged T-cell priming when Ag-donor DCs could not present Ag.


Asunto(s)
Antígenos/metabolismo , Linfocitos T CD8-positivos/inmunología , Reactividad Cruzada , Células Dendríticas/inmunología , Ganglios Linfáticos/inmunología , Animales , Presentación de Antígeno , Antígenos/inmunología , Antígenos CD/genética , Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos T/genética , Antígenos de Diferenciación de Linfocitos T/inmunología , Linfocitos T CD8-positivos/metabolismo , Movimiento Celular , Células Dendríticas/química , Células Dendríticas/metabolismo , Microscopía Intravital , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Ganglios Linfáticos/citología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/genética , Ovalbúmina/inmunología , Virus Vaccinia/genética , Virus Vaccinia/fisiología
4.
Nat Cancer ; 5(5): 742-759, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38429414

RESUMEN

Successful immunotherapy relies on triggering complex responses involving T cell dynamics in tumors and the periphery. Characterizing these responses remains challenging using static human single-cell atlases or mouse models. To address this, we developed a framework for in vivo tracking of tumor-specific CD8+ T cells over time and at single-cell resolution. Our tools facilitate the modeling of gene program dynamics in the tumor microenvironment (TME) and the tumor-draining lymph node (tdLN). Using this approach, we characterize two modes of anti-programmed cell death protein 1 (PD-1) activity, decoupling induced differentiation of tumor-specific activated precursor cells from conventional type 1 dendritic cell (cDC1)-dependent proliferation and recruitment to the TME. We demonstrate that combining anti-PD-1 therapy with anti-4-1BB agonist enhances the recruitment and proliferation of activated precursors, resulting in tumor control. These data suggest that effective response to anti-PD-1 therapy is dependent on sufficient influx of activated precursor CD8+ cells to the TME and highlight the importance of understanding system-level dynamics in optimizing immunotherapies.


Asunto(s)
Linfocitos T CD8-positivos , Inmunoterapia , Microambiente Tumoral , Animales , Ratones , Inmunoterapia/métodos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Microambiente Tumoral/inmunología , Humanos , Neoplasias/inmunología , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Células Dendríticas/inmunología , Células Dendríticas/efectos de los fármacos , Línea Celular Tumoral
5.
Sci Immunol ; 8(81): eadd8005, 2023 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-36867679

RESUMEN

FDA-approved anti-PD-L1 monoclonal antibodies (mAbs) bear the IgG1 isotype, whose scaffolds are either wild-type (e.g., avelumab) or Fc-mutated and lacking Fcγ receptor (FcγR) engagement (e.g., atezolizumab). It is unknown whether variation in the ability of the IgG1 Fc region to engage FcγRs renders mAbs with superior therapeutic activity. In this study, we used humanized FcγR mice to study the contribution of FcγR signaling to the antitumor activity of human anti-PD-L1 mAbs and to identify an optimal human IgG scaffold for PD-L1 mAbs. We observed similar antitumor efficacy and comparable tumor immune responses in mice treated with anti-PD-L1 mAbs with wild-type and Fc-mutated IgG scaffolds. However, in vivo antitumor activity of the wild-type anti-PD-L1 mAb avelumab was enhanced by combination treatment with an FcγRIIB-blocking antibody, which was co-administered to overcome the suppressor function of FcγRIIB in the tumor microenvironment (TME). We performed Fc glycoengineering to remove the fucose subunit from the Fc-attached glycan of avelumab to enhance its binding to the activating FcγRIIIA. Treatment with the Fc-afucosylated version of avelumab also enhanced antitumor activity and induced stronger antitumor immune responses compared with the parental IgG. The enhanced effect by afucosylated PD-L1 antibody was dependent on neutrophils and associated with decreased frequencies of PD-L1+ myeloid cells and increased infiltration of T cells in the TME. Our data reveal that the current design of FDA-approved anti-PD-L1 mAbs does not optimally harness FcγR pathways and suggest two strategies to enhance FcγR engagement to optimize anti-PD-L1 immunotherapy.


Asunto(s)
Anticuerpos Monoclonales , Receptores de IgG , Humanos , Animales , Ratones , Inmunoglobulina G , Inhibidores de Puntos de Control Inmunológico , Inmunoterapia
6.
Nat Cancer ; 3(3): 287-302, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35190724

RESUMEN

Therapeutic use of agonistic anti-CD40 antibodies is a potentially powerful approach for activation of the immune response to eradicate tumors. However, the translation of this approach to clinical practice has been substantially restricted due to the severe dose-limiting toxicities observed in multiple clinical trials. Here, we demonstrate that conventional type 1 dendritic cells are essential for triggering antitumor immunity but not the toxicity of CD40 agonists, while macrophages, platelets and monocytes lead to toxic events. Therefore, we designed bispecific antibodies that target CD40 activation preferentially to dendritic cells, by coupling the CD40 agonist arm with CD11c-, DEC-205- or CLEC9A-targeting arms. These bispecific reagents demonstrate a superior safety profile compared to their parental CD40 monospecific antibody while triggering potent antitumor activity. We suggest such cell-selective bispecific agonistic antibodies as a drug platform to bypass the dose-limiting toxicities of anti-CD40, and of additional types of agonistic antibodies used for cancer immunotherapy.


Asunto(s)
Anticuerpos Biespecíficos , Neoplasias , Anticuerpos Biespecíficos/farmacología , Antígenos CD40 , Células Dendríticas , Humanos , Inmunoterapia , Neoplasias/tratamiento farmacológico
7.
J Autoimmun ; 36(1): 16-24, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21035305

RESUMEN

The autoimmune regulator (Aire) is involved in the prevention of autoimmunity by promoting thymic expression of tissue restricted antigens which leads to elimination of self-reactive T cells. We found that Aire knockout (KO) mice as well as mouse strains that are susceptible to experimental autoimmune myasthenia gravis (EAMG) have lower thymic expression of acetylcholine receptor (AChR- the main autoantigen in MG), compared to wild type (WT) mice and EAMG-resistant mouse strains, respectively. We demonstrated that Aire KO mice have a significant and reproducible lower frequency of CD4+Foxp3+ cells and a higher expression of Th17 markers in their thymus, compared to wild type (WT) mice. These findings led us to expect that Aire KO mice would display increased susceptibility to EAMG. Surprisingly, when EAMG was induced in young (2 month-old) mice, EAMG was milder in Aire KO than in WT mice for several weeks until the age of about 5 months. However, when EAMG was induced in relatively aged (6 month-old) mice, Aire KO mice presented higher disease severity than WT controls. This age-related change in susceptibility to EAMG correlated with an elevated proportion of Treg cells in the spleens of young but not old KO, compared to WT mice, suggesting a role for peripheral Treg cells in the course of disease. Our observations point to a possible link between Aire and Treg cells and suggest an involvement for both in the pathogenesis of myasthenia.


Asunto(s)
Miastenia Gravis Autoinmune Experimental/inmunología , Linfocitos T Reguladores/inmunología , Timo/inmunología , Factores de Transcripción/inmunología , Factores de Edad , Animales , Separación Celular , Susceptibilidad a Enfermedades/inmunología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Colinérgicos/inmunología , Receptores Colinérgicos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/citología , Bazo/inmunología , Timo/citología , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Proteína AIRE
8.
Cell Rep ; 37(11): 110114, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34883043

RESUMEN

Messenger RNA-based vaccines against COVID-19 induce a robust anti-SARS-CoV-2 antibody response with potent viral neutralization activity. Antibody effector functions are determined by their constant region subclasses and by their glycosylation patterns, but their role in vaccine efficacy is unclear. Moreover, whether vaccination induces antibodies similar to those in patients with COVID-19 remains unknown. We analyze BNT162b2 vaccine-induced IgG subclass distribution and Fc glycosylation patterns and their potential to drive effector function via Fcγ receptors and complement pathways. We identify unique and dynamic pro-inflammatory Fc compositions that are distinct from those in patients with COVID-19 and convalescents. Vaccine-induced anti-Spike IgG is characterized by distinct Fab- and Fc-mediated functions between different age groups and in comparison to antibodies generated during natural viral infection. These data highlight the heterogeneity of Fc responses to SARS-CoV-2 infection and vaccination and suggest that they support long-lasting protection differently.


Asunto(s)
COVID-19/inmunología , Glicosilación/efectos de los fármacos , SARS-CoV-2/inmunología , Adulto , Anciano , Anticuerpos Antivirales/inmunología , Vacuna BNT162/inmunología , Vacunas contra la COVID-19/metabolismo , Femenino , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Israel/epidemiología , Masculino , Persona de Mediana Edad , Glicoproteína de la Espiga del Coronavirus/inmunología , Vacunación/métodos , Eficacia de las Vacunas , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/metabolismo , Vacunas de ARNm/inmunología , Vacunas de ARNm/metabolismo
9.
J Neuroimmunol ; 345: 577281, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32534388

RESUMEN

To identify the mechanisms relevant for the therapeutic effect of glatiramer acetate (GA), we studied T- and B- regulatory cells as well as GM-CSF expression in mice recovered from experimental autoimmune encephalomyelitis (EAE). Selective depletion of Tregs reduced but did not eliminate the ability of GA to ameliorate EAE, indicating a role for additional immune-subsets. The prevalence of Bregs in the periphery and the CNS of EAE-mice increased following GA-treatment. Furthermore, GA downregulated the pathological expression of GM-CSF, on both the protein and mRNA levels. These findings corroborate the broad immunomodulatory mechanism of action of GA in EAE/MS.


Asunto(s)
Linfocitos B Reguladores/metabolismo , Acetato de Glatiramer/farmacología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Inmunosupresores/farmacología , Esclerosis Múltiple/metabolismo , Linfocitos T Reguladores/metabolismo , Animales , Linfocitos B Reguladores/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Acetato de Glatiramer/uso terapéutico , Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Inmunosupresores/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Esclerosis Múltiple/tratamiento farmacológico , Linfocitos T Reguladores/efectos de los fármacos
10.
PLoS One ; 15(11): e0241164, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33227020

RESUMEN

The COVID-19 pandemic and the fast global spread of the disease resulted in unprecedented decline in world trade and travel. A critical priority is, therefore, to quickly develop serological diagnostic capacity and identify individuals with past exposure to SARS-CoV-2. In this study serum samples obtained from 309 persons infected by SARS-CoV-2 and 324 of healthy, uninfected individuals as well as serum from 7 COVID-19 patients with 4-7 samples each ranging between 1-92 days post first positive PCR were tested by an "in house" ELISA which detects IgM, IgA and IgG antibodies against the receptor binding domain (RBD) of SARS-CoV-2. Sensitivity of 47%, 80% and 88% and specificity of 100%, 98% and 98% in detection of IgM, IgA and IgG antibodies, respectively, were observed. IgG antibody levels against the RBD were demonstrated to be up regulated between 1-7 days after COVID-19 detection, earlier than both IgM and IgA antibodies. Study of the antibody kinetics of seven COVID 19 patients revealed that while IgG levels are high and maintained for at least 3 months, IgM and IgA levels decline after a 35-50 days following infection. Altogether, these results highlight the usefulness of the RBD based ELISA, which is both easy and cheap to prepare, to identify COVID-19 patients even at the acute phase. Most importantly our results demonstrate that measuring IgG levels alone is both sufficient and necessary to diagnose past exposure to SARS-CoV-2.


Asunto(s)
Anticuerpos Antivirales/inmunología , Prueba Serológica para COVID-19/métodos , COVID-19/diagnóstico , COVID-19/epidemiología , Inmunoglobulina G/inmunología , Pandemias , Dominios Proteicos/inmunología , SARS-CoV-2/inmunología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , COVID-19/sangre , COVID-19/virología , Estudios de Cohortes , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunoglobulina A/sangre , Inmunoglobulina A/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Inmunoglobulina M/inmunología , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , SARS-CoV-2/genética , Sensibilidad y Especificidad , Adulto Joven
11.
J Neuroimmunol ; 201-202: 128-35, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18632163

RESUMEN

Myasthenia gravis (MG) is frequently treated by corticosteroids such as methylprednisolone. However, continuous treatment with steroids often results in adverse effects. In the present study we evaluated the therapeutic potential of a combination of suboptimal doses of methylprednisolone (Solumedrol) and Pentoxifylline (PTX), a general phosphodiesterase (PDE) inhibitor, in rat experimental autoimmune MG (EAMG). This combined treatment resulted in a pronounced suppressive effect on EAMG and was by far more effective than each of the drugs administered separately at these low doses. The suppressive effect on EAMG was accompanied by decreased humoral and cellular responses to AChR as well as down-regulated mRNA expression levels of Th1 cytokines and IL-10 in lymph node cells and of PDE-4 and cathepsin-l in the muscle. This study demonstrates the potential of PTX as a steroid-sparing agent in the management of myasthenia gravis.


Asunto(s)
Glucocorticoides/uso terapéutico , Metilprednisolona/uso terapéutico , Miastenia Gravis Autoinmune Experimental/tratamiento farmacológico , Pentoxifilina/uso terapéutico , Inhibidores de Fosfodiesterasa/uso terapéutico , Animales , Anticuerpos/sangre , Peso Corporal/efectos de los fármacos , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Miastenia Gravis Autoinmune Experimental/sangre , Miastenia Gravis Autoinmune Experimental/patología , Ratas , Ratas Endogámicas Lew , Receptores Colinérgicos/inmunología , Índice de Severidad de la Enfermedad
12.
J Neuroimmunol ; 194(1-2): 89-96, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18178258

RESUMEN

Intravenous immunoglobulin (IVIG) treatment is beneficially used in autoimmune disorders including myasthenia gravis (MG) although its mode of action and active components are still not fully identified. In an attempt to isolate from IVIG a disease-specific suppressive fraction, IVIG was passed on columns of IgG from rats with experimental autoimmune MG (EAMG) or from MG patients. These chromatographies resulted in depletion of the suppressive activity of IVIG on rat EAMG whereas the minute amounts of IgG fractions eluted from the EAMG- or MG-specific columns retained the immunosuppressive activity of IVIG. These results demonstrate that a minor disease-specific immunoglobulin fraction present in IVIG is essential for its suppressive activity.


Asunto(s)
Anticuerpos Antiidiotipos/aislamiento & purificación , Inmunoglobulinas Intravenosas/uso terapéutico , Inmunosupresores/uso terapéutico , Miastenia Gravis Autoinmune Experimental/terapia , Animales , Anticuerpos Antiidiotipos/inmunología , Anticuerpos Antiidiotipos/uso terapéutico , Especificidad de Anticuerpos , Autoanticuerpos/inmunología , Cromatografía de Afinidad , Adyuvante de Freund/inmunología , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulinas Intravenosas/química , Técnicas de Inmunoadsorción , Inmunosupresores/aislamiento & purificación , Miastenia Gravis/inmunología , Miastenia Gravis Autoinmune Experimental/inmunología , Ratas , Especificidad de la Especie
13.
Ann N Y Acad Sci ; 1132: 244-8, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18567875

RESUMEN

Intravenous immunoglobulin (IVIG) administration has been beneficially used for the treatment of a variety of autoimmune diseases including myasthenia gravis (MG). We have demonstrated that IVIG administration in experimental autoimmune MG (EAMG) results in suppression of disease that is accompanied by decreased Th1 cell and B cell proliferation. Chromatography of pooled human immunoglobulins (IVIG) on immobilized IgG, isolated from rats with EAMG or from MG patients, results in a depletion of the suppressive activity of the IVIG. Moreover, reconstitution of the activity-depleted IVIG with the eluted minute IVIG fractions that had been adsorbed onto the EAMG- or MG-specific columns recovers the depleted immunosuppressive activity. This study supports the notion that the therapeutic effect of IVIG is mediated by an antigen-specific anti-immunoglobulin (anti-idiotypic) activity that is essential for its suppressive activity.


Asunto(s)
Especificidad de Anticuerpos/inmunología , Inmunoglobulinas Intravenosas/inmunología , Inmunoglobulinas Intravenosas/uso terapéutico , Inmunosupresores/inmunología , Inmunosupresores/uso terapéutico , Miastenia Gravis Autoinmune Experimental/inmunología , Miastenia Gravis Autoinmune Experimental/terapia , Animales , Humanos , Inmunoterapia , Ratas , Receptores Colinérgicos/inmunología
14.
Isr Med Assoc J ; 10(1): 58-60, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18300576

RESUMEN

BACKGROUND: [corrected] Intravenous immunoglobulin administration has been beneficially used for the treatment of a variety of autoimmune diseases including myasthenia gravis, although its mode of action and active components have not yet been fully identified. OBJECTIVES: To isolate from IVIg a disease-specific fraction involved in the therapeutic activity in myasthenia and to identify its properties and function. RESULTS: IVIg administration in experimental autoimmune MG results in suppression of disease that is accompanied by decreased Th1 cell and B cell proliferation. Chromatography of IVIg on columns of IgG from rats with EAMG or from MG patients resulted in depletion of the suppressive activity that IVIg has on rat EAMG. Moreover, the minute amounts of IgG fractions eluted from the EAMG or MG-specific columns retained the immunosuppressive activity of IVIg. CONCLUSIONS: Our study supports the notion that the therapeutic effect of IVIg is mediated by a minor disease-specific immunoglobulin fraction that is present in IVIg and is essential for its therapeutic activity.


Asunto(s)
Inmunoglobulinas Intravenosas/farmacología , Factores Inmunológicos/farmacología , Miastenia Gravis Autoinmune Experimental/tratamiento farmacológico , Animales , Linfocitos B/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Fraccionamiento Químico , Cromatografía , Humanos , Ratas , Células TH1/efectos de los fármacos
15.
Stem Cells Transl Med ; 7(1): 68-77, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29266820

RESUMEN

Repair of injured lungs represents a longstanding therapeutic challenge. We recently demonstrated that human and mouse embryonic lung tissue from the canalicular stage of development are enriched with lung progenitors, and that a single cell suspension of canalicular lungs can be used for transplantation, provided that lung progenitor niches in the recipient mice are vacated by strategies similar to those used in bone marrow transplantation. Considering the ethical limitations associated with the use of fetal cells, we investigated here whether adult lungs could offer an alternative source of lung progenitors for transplantation. We show that intravenous infusion of a single cell suspension of adult mouse lungs from GFP+ donors, following conditioning of recipient mice with naphthalene and subsequent sublethal irradiation, led to marked colonization of the recipient lungs, at 6-8 weeks post-transplant, with donor derived structures including epithelial, endothelial, and mesenchymal cells. Epithelial cells within these donor-derived colonies expressed markers of functionally distinct lung cell types, and lung function, which is significantly compromised in mice treated with naphthalene and radiation, was found to be corrected following transplantation. Dose response analysis suggests that the frequency of patch forming cells in adult lungs was about threefold lower compared to that found in E16 fetal lungs. However, as adult lungs are much larger, the total number of patch forming cells that can be collected from this source is significantly greater. Our study provides proof of concept for lung regeneration by adult lung cells after preconditioning to vacate the pulmonary niche. Stem Cells Translational Medicine 2018;7:68-77.


Asunto(s)
Células Epiteliales/trasplante , Regeneración Tisular Dirigida/métodos , Lesión Pulmonar/terapia , Pulmón/citología , Trasplante de Células Madre , Células Madre/citología , Animales , Células Cultivadas , Células Epiteliales/citología , Femenino , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Naftalenos/toxicidad
16.
Cancer Discov ; 8(11): 1366-1375, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30209080

RESUMEN

The quest for tumor-associated antigens (TAA) and neoantigens is a major focus of cancer immunotherapy. Here, we combine a neoantigen prediction pipeline and human leukocyte antigen (HLA) peptidomics to identify TAAs and neoantigens in 16 tumors derived from seven patients with melanoma and characterize their interactions with their tumor-infiltrating lymphocytes (TIL). Our investigation of the antigenic and T-cell landscapes encompassing the TAA and neoantigen signatures, their immune reactivity, and their corresponding T-cell identities provides the first comprehensive analysis of cancer cell T-cell cosignatures, allowing us to discover remarkable antigenic and TIL similarities between metastases from the same patient. Furthermore, we reveal that two neoantigen-specific clonotypes killed 90% of autologous melanoma cells, both in vitro and in vivo, showing that a limited set of neoantigen-specific T cells may play a central role in melanoma tumor rejection. Our findings indicate that combining HLA peptidomics with neoantigen predictions allows robust identification of targetable neoantigens, which could successfully guide personalized cancer immunotherapies.Significance: As neoantigen targeting is becoming more established as a powerful therapeutic approach, investigating these molecules has taken center stage. Here, we show that a limited set of neoantigen-specific T cells mediates tumor rejection, suggesting that identifying just a few antigens and their corresponding T-cell clones could guide personalized immunotherapy. Cancer Discov; 8(11); 1366-75. ©2018 AACR. This article is highlighted in the In This Issue feature, p. 1333.


Asunto(s)
Presentación de Antígeno/inmunología , Antígenos de Neoplasias/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma/inmunología , Linfocitos T/inmunología , Animales , Antígenos de Neoplasias/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Linfocitos T/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
17.
FASEB J ; 20(2): 374-6, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16365386

RESUMEN

Myasthenia gravis (MG) and experimental autoimmune MG (EAMG) are T cell-dependent antibody-mediated autoimmune disorders, in which the nicotinic acetylcholine receptor (AChR) is the major autoantigen. DNA microarray analysis revealed increased levels of several phosphodiesterase (PDE) subtypes in lymph node cells (LNC) and muscles of EAMG rats compared with healthy controls. Quantitative real-time PCR analysis indicated that EAMG is characterized by an increase of PDE subtypes 1, 3, 4, and 7 in LNC and of PDE subtypes 2, 3, 4, and 7 in muscles. Pentoxifylline (PTX), a general PDE inhibitor, inhibited the progression of EAMG when treatment started at either the acute or chronic stages of disease. This suppression was associated with down-regulation of humoral and cellular AChR-specific responses, as well as down-regulation of PDE4, TNF-alpha, IL-18, IL-12, and IL-10 in LNC and of PDEs 1, 4, 7, and TNF-alpha in muscles. The expression of Foxp3, a transcription factor essential for CD4+CD25+ regulatory T cell function, was increased in splenocytes although the number of these cells remained unchanged. PTX also reduced the expression of the endopeptidase cathepsin-l, a marker of muscle damage, in EAMG muscles. This study demonstrates the involvement of PDE regulation in EAMG pathogenesis and suggests that PDE inhibitors may be considered for immunotherapy of MG.


Asunto(s)
Miastenia Gravis/tratamiento farmacológico , Miastenia Gravis/enzimología , Pentoxifilina/farmacología , Inhibidores de Fosfodiesterasa/farmacología , Hidrolasas Diéster Fosfóricas/metabolismo , Animales , Catepsinas/metabolismo , Línea Celular , Citocinas/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Humanos , Hidrolasas Diéster Fosfóricas/genética , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas Lew , Receptores Colinérgicos/metabolismo , Linfocitos T Reguladores/metabolismo
18.
Ann N Y Acad Sci ; 1107: 111-7, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17804538

RESUMEN

DNA microarray technology was used to identify new potential drug targets for myasthenia gravis (MG), to delineate genes involved in the pathogenesis of the disease and to possibly target their protein products for immunotherapy. In this study we compared the gene expression in lymph node cells (LNC) and muscles of rats with experimental autoimmune MG (EAMG) to those of control, healthy rats. Of the genes that were found to be deregulated in EAMG, we chose to elaborate on two gene systems: (a) The chemokine IFN-gamma-inducible protein 10 (IP-10, CXCL10), and its receptor (CXCR3) and (b) phosphodiesterases.


Asunto(s)
Evaluación Preclínica de Medicamentos , Miastenia Gravis/tratamiento farmacológico , Miastenia Gravis/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Corticoesteroides/uso terapéutico , Animales , Quimiocina CXCL10 , Quimiocinas CXC/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Miastenia Gravis/metabolismo , Miastenia Gravis/patología , Inhibidores de Fosfodiesterasa/uso terapéutico , Receptores CXCR3 , Receptores de Quimiocina/genética
19.
Ann N Y Acad Sci ; 1110: 550-8, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17911471

RESUMEN

Intravenous immunoglobulin (IVIG) administration has been beneficially used for the treatment of a variety of autoimmune diseases including myasthenia gravis (MG). We have demonstrated that IVIG administration in experimental autoimmune MG (EAMG) results in suppression of disease that is accompanied by decreased Th1 cell and B cell proliferation. Chromatography of pooled human immunoglobulins (IVIGs) on immobilized IgG, isolated from rats with EAMG, results in a complete depletion of the suppressive activity of the IVIG. Moreover, the eluate from this EAMG-specific antibody column retains the immunosuppressive activity of IVIG. This study supports the notion that the therapeutic effect of IVIGs is mediated by an antigen-specific anti-immunoglobulin (anti-idiotypic) activity that is essential for its suppressive activity.


Asunto(s)
Inmunoglobulina G/aislamiento & purificación , Inmunoglobulina G/uso terapéutico , Inmunoglobulinas Intravenosas/inmunología , Inmunoglobulinas Intravenosas/uso terapéutico , Miastenia Gravis Autoinmune Experimental/inmunología , Miastenia Gravis Autoinmune Experimental/terapia , Animales , Humanos , Inmunoglobulina G/inmunología , Inmunoterapia , Miastenia Gravis Autoinmune Experimental/patología , Ratas
20.
Cell Rep ; 20(11): 2547-2555, 2017 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-28903036

RESUMEN

Cytotoxic T lymphocytes (CTLs) used in immunotherapy are typically cultured under atmospheric O2 pressure but encounter hypoxic conditions inside tumors. Activating CTLs under hypoxic conditions has been shown to improve their cytotoxicity in vitro, but the mechanism employed and the implications for immunotherapy remain unknown. We activated and cultured OT-I CD8 T cells at either 1% or 20% O2. Hypoxic CTLs survived, as well as normoxic ones, in vitro but killed OVA-expressing B16 melanoma cells more efficiently. Hypoxic CTLs contained similar numbers of cytolytic granules and released them as efficiently but packaged more granzyme-B in each granule without producing more perforin. We imaged CTL distribution and motility inside B16-OVA tumors using confocal and intravital 2-photon microscopy and observed no obvious differences. However, mice treated with hypoxic CTLs exhibited better tumor regression and survived longer. Thus, hypoxic CTLs may perform better in tumor immunotherapy because of higher intrinsic cytotoxicity rather than improved migration inside tumors.


Asunto(s)
Antineoplásicos/metabolismo , Citotoxicidad Inmunológica , Neoplasias/inmunología , Neoplasias/patología , Linfocitos T Citotóxicos/inmunología , Animales , Diferenciación Celular , Hipoxia de la Célula/inmunología , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Granzimas/metabolismo , Ratones , Neoplasias/irrigación sanguínea , Análisis de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA