Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Front Immunol ; 15: 1380065, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38726005

RESUMEN

Introduction: Solid cancers Myeloid cells are prevalent in solid cancers, but they frequently exhibit an anti-inflammatory pro-tumor phenotype that contribute to the immunosuppressive tumor microenvironment (TME), which hinders the effectiveness of cancer immunotherapies. Myeloid cells' natural ability of tumor trafficking makes engineered myeloid cell therapy an intriguing approach to tackle the challenges posed by solid cancers, including tumor infiltration, tumor cell heterogenicity and the immunosuppressive TME. One such engineering approach is to target the checkpoint molecule PD-L1, which is often upregulated by solid cancers to evade immune responses. Method: Here we devised an adoptive cell therapy strategy based on myeloid cells expressing a Chimeric Antigen Receptor (CAR)-like immune receptor (CARIR). The extracellular domain of CARIR is derived from the natural inhibitory receptor PD-1, while the intracellular domain(s) are derived from CD40 and/or CD3ζ. To assess the efficacy of CARIR-engineered myeloid cells, we conducted proof-of-principle experiments using co-culture and flow cytometry-based phagocytosis assays in vitro. Additionally, we employed a fully immune-competent syngeneic tumor mouse model to evaluate the strategy's effectiveness in vivo. Result: Co-culturing CARIR-expressing human monocytic THP-1 cells with PD-L1 expressing target cells lead to upregulation of the costimulatory molecule CD86 along with expression of proinflammatory cytokines TNF-1α and IL-1ß. Moreover, CARIR expression significantly enhanced phagocytosis of multiple PD-L1 expressing cancer cell lines in vitro. Similar outcomes were observed with CARIR-expressing human primary macrophages. In experiments conducted in syngeneic BALB/c mice bearing 4T1 mammary tumors, infusing murine myeloid cells that express a murine version of CARIR significantly slowed tumor growth and prolonged survival. Conclusion: Taken together, these results demonstrate that adoptive transfer of PD-1 CARIR-engineered myeloid cells represents a promising strategy for treating PD-L1 positive solid cancers.


Asunto(s)
Antígeno B7-H1 , Inmunoterapia Adoptiva , Células Mieloides , Receptores Quiméricos de Antígenos , Microambiente Tumoral , Animales , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Ratones , Humanos , Células Mieloides/inmunología , Células Mieloides/metabolismo , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Inmunoterapia Adoptiva/métodos , Microambiente Tumoral/inmunología , Línea Celular Tumoral , Femenino , Neoplasias/inmunología , Neoplasias/terapia
2.
Spectrochim Acta A Mol Biomol Spectrosc ; 316: 124328, 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-38669986

RESUMEN

We designed and developed the probe W-3 for detection of Cu2+. The results showed probe can selectively detect Cu2+, accompanied by noticeable color change. The probe can detect the Cu2+ in water samples and drinks based on absorption detection. In addition, the combination of portable test paper and the smartphone platform obtained great convenience for on-site and visual detection of Cu2+, with satisfactory sensitivity and reliability. More importantly, the fluorescence probe W-3 can be used for the detection of Cu2+ in cells and mice. Therefore, the W-3 provided potential chemical tools for detecting Cu2+ in vitro and vivo.


Asunto(s)
Cobre , Colorantes Fluorescentes , Espectrometría de Fluorescencia , Cobre/análisis , Colorantes Fluorescentes/química , Animales , Espectrometría de Fluorescencia/métodos , Humanos , Ratones , Imagen Óptica/métodos , Células HeLa , Límite de Detección
3.
Biosens Bioelectron ; 254: 116233, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38518563

RESUMEN

Intracellular microenvironment (viscosity and polarity) and peroxynitrite ions (ONOO-) are involved in maintaining cell morphology, cell function, and signaling so that it is crucial to explore their level changes in vitro and vivo. In this work, we designed and synthesized a mitochondria-targeted fluorescence probe XBL for monitoring the dynamic changes of viscosity, polarity, and ONOO- based on TICT and ICT mechanism. The fluorescence spectra showed obvious changes for polarity at 500 nm as well as ONOO- and viscosity at 660 nm, respectively. The XBL can image simultaneously viscosity, polarity, and ONOO- in cells, and the results showed excess ONOO- leaded to the increase of viscosity in mitochondrial. The ferroptosis process was accompanied by increase of intracellular viscosity and ONOO- levels (or decrease of polarity), which allowed us to better understand the relevant physiological and pathological processes. The XBL can distinguish normal cells and cancerous cells by the fluorescence intensity changes in green and red channels, and image viscosity in inflamed mice. Thus, XBL can provided the chemical tool to understand the physiological and pathological mechanisms of disease by simultaneous detection of viscosity, polarity and ONOO-.


Asunto(s)
Técnicas Biosensibles , Colorantes Fluorescentes , Ratones , Animales , Viscosidad , Células RAW 264.7 , Mitocondrias , Ácido Peroxinitroso
4.
Front Oncol ; 13: 1244545, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37637071

RESUMEN

Objective: This study aimed to provide a realistic observation of survival by major site for 48,866 cancer patients treated at a tertiary cancer hospital in a rural area of China. Methods: Patients with cancer registered between 2007 and 2017 in the Nantong rural area were followed up. The starting date for survival calculation was the date of the first diagnosis of cancer at the Nantong Tumor Hospital, and the closing date was December 31, 2020. Observed survival (OS) was analyzed according to ICD-10 site, sex, age, region, and hospitalization period using the life table method and compared using the Wilcoxon (Gehan) statistic. Results: The overall 5-year OS rate was 40.48% for all 48,866 patients, 30.19% for males, and 51.90% for females. The top five cancer sites, accounting for 60.51% of the total cases, were the esophagus, lung, stomach, liver, and cervix, with 5-year OS rates of 33.72%, 18.64%, 32.10%, 19.04%, and 71.51%, respectively. The highest 5-year OS was observed in the thyroid (87.52%) and the lowest was in the pancreas (6.37%). Survival was significantly higher in younger patients than in older patients, with 5-year OSs of 69.26% and 19.84% in those aged 20-29 and 90-99 years, respectively. Five-year OSs improved significantly from 39.35% in 2007-2011 to 41.26% in 2012-2017. Conclusion: Overall survival improved over the years, although the improvement at some sites was not significant. The observed survival varies from region to region, reflecting differences in the patterns of major sites, disparities in proportions of hospitalization, and demographic characteristics.

5.
Cancer J ; 28(4): 257-262, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35880934

RESUMEN

ABSTRACT: Adoptive cellular therapies are making major strides in the treatment of cancer, both for hematologic and solid tumors. These cellular products include chimeric antigen receptor T cells and T-cell receptor-modified T cells, tumor-infiltrating lymphocytes, marrow-infiltrating T cells, natural killer cells as well as macrophage-based therapeutics. Advancement in genomics, computational biology, immunology, and cell therapy manufacturing has facilitated advancement of adoptive T cell therapies into the clinic, whereas clinical efficacy has driven Food and Drug Administration approvals. The growth of adoptive cellular therapy has, in turn, led to innovation in the preclinical models available, from ex vivo cell-based models to in vivo xenograft models of treatment. This review focuses on the development and application of in vitro models and in vivo models (cell line xenograft, humanized mice, and patient-derived xenograft models) that directly evaluate these human cellular products.


Asunto(s)
Inmunoterapia Adoptiva , Neoplasias , Animales , Línea Celular Tumoral , Tratamiento Basado en Trasplante de Células y Tejidos , Humanos , Ratones , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T
6.
Cell Immunol ; 375: 104515, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35417812

RESUMEN

Multiple sclerosis disproportionally affects women. The present study was undertaken to determine whether NFAT5 contributed to the pathogenesis of experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis, and if it did, whether the impact was sex associated. NFAT5 haplodeficiency reduced the disease severity only in female mice. This effect was associated with significant increases in frequency of T regulatory (Treg) cells in the CNS (from 1.45 ± 0.39% to 3.73 ± 0.94%) and spleen from (0.31 ± 0.06% to 0.94 ± 0.29%) without significantly affecting the CNS CD4+ subsets frequency. NFAT5 haploinsufficiency also significantly reduced the frequency of CD11c+CD8α+ dendritic cells in the female CNS. However, increase of their frequency in the CNS via intraperitoneal Flt3L injection at peak EAE had no significant effect on the disease courses. We conclude that NFAT5 contributes to pathogenesis of EAE in female mice, possibly through decreasing tissue specific frequency of Treg cells.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Linfocitos T Reguladores , Factores de Transcripción , Animales , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple , Bazo , Factores de Transcripción/genética
7.
Clin Exp Rheumatol ; 40(3): 560-567, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33822701

RESUMEN

OBJECTIVES: No approved pharmacotherapies are available for patients with interstitial pneumonia with autoimmune features (IPAF). In the present work, we aimed to evaluate the efficacy and safety of pirfenidone for the treatment of IPAF. METHODS: A retrospective cohort study consisting of patients who met diagnostic criteria for IPAF was performed after a multidisciplinary review, and the patients receiving pirfenidone were compared with those in the non-pirfenidone group. The baseline data and diagnostic characteristics of patients were assessed. Pulmonary function and prednisone dose were analysed by a mix-effects model. RESULTS: A total of 184 patients, who met the diagnostic criteria of IPAF, were divided into two groups: pirfenidone group (n=81) and non-pirfenidone group (n=103). Patients in the pirfenidone group had a lower forced vital capacity (FVC%, p<0.001) and a lower diffusion capacity for carbon monoxide (DLCO%, p=0.003). The pirfenidone group exhibited a greater increase of FVC% at 6 (p=0.003), 12 (p=0.013), and 24 (p=0.003) months. After adjustment for sex, age, UIP pattern, baseline FVC% and DLCO%, patients in the pirfenidone group continued to show a greater improvement in FVC% (χ2(1)=4.59, p=0.032). Subgroup analysis identified superior therapeutic effects of pirfenidone in patients with dosage >600 mg/day (p=0.010) and medication course >12 months (p=0.007). Besides, the pirfenidone group had a lower prednisone dose than the non-pirfenidone group after 12 months of treatment (p=0.002). Moreover, 17 patients (19.32%) experienced side effects after taking pirfenidone, including one case of anaphylactic shock. CONCLUSIONS: Pirfenidone (600-1,800 mg/day) might help improve FVC, with an acceptable safety and tolerability profile in IPAF patients.


Asunto(s)
Fibrosis Pulmonar Idiopática , Enfermedades Pulmonares Intersticiales , Humanos , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Enfermedades Pulmonares Intersticiales/diagnóstico , Piridonas/efectos adversos , Estudios Retrospectivos , Capacidad Vital
8.
Cell Immunol ; 358: 104222, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33053469

RESUMEN

The expansion of polyclonal T regulatory cells (Tregs) offers great promise for the treatment of immune-mediated diseases, such as multiple sclerosis (MS). However, polyclonal Tregs can be non-specifically immunosuppressive. Based on the advancements with chimeric antigen receptor (CAR) therapy in leukemia, we previously engineered Tregs to express a T-cell receptor (TCR) specific for a myelin basic protein (MBP) peptide. These TCR-engineered specific Tregs suppressed the proliferation of MBP-reactive T effector cells and ameliorated myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE). Herein, we extend this approach by creating human regulatory T cells expressing functional single-chain chimeric antigen receptors (scFv CAR), targeting either MBP or MOG. These scFv CAR-transduced Tregs retained FoxP3 and Helios, characteristic of Treg cells, after long-term expansion in vitro. Importantly, these engineered CNS targeting CAR-Tregs were able to suppress autoimmune pathology in EAE, demonstrating that these Tregs have the potential to be used as a cellular therapy for MS patients.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Proteína Básica de Mielina/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Encefalomielitis Autoinmune Experimental/terapia , Femenino , Factores de Transcripción Forkhead/metabolismo , Humanos , Tolerancia Inmunológica/inmunología , Inmunoterapia Adoptiva/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Esclerosis Múltiple/inmunología , Proteína Básica de Mielina/inmunología , Vaina de Mielina/metabolismo , Vaina de Mielina/patología , Glicoproteína Mielina-Oligodendrócito/inmunología , Ingeniería de Proteínas/métodos , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo
9.
Front Immunol ; 11: 693, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32373126

RESUMEN

Anti-drug antibody formation poses tremendous obstacles for optimal treatment of hemophilia A (HA). In this study, we sought to utilize chimeric receptor-modified natural regulatory T cells (Tregs) to target FVIII-specific memory B cells, which are responsible for persistent anti-FVIII neutralizing antibodies (inhibitors) in HA patients. Thus, CD4+CD25 hi CD304+ natural Tregs were FACS sorted from naïve C57BL/6 mice and retrovirally transduced to express a chimeric B-cell antibody receptor (BAR) containing the immunodominant A2 domain of FVIII. Plasmablast-depleted (CD138 neg ) splenocytes from FVIII immunized FVIII-knockout HA mice served as the source for FVIII-specific memory B cells, which were specifically stimulated in vitro with FVIII and enumerated in a B-cell ELISPOT assays. Adding A2-BAR Tregs (1 per 150 splenocytes), but not conventional T cells, to the CD138- splenocytes significantly suppressed the formation of anti-FVIII antibody secreting cells (ASC), compared to the non-relevant OVA-BAR Tregs control group. The observation that A2-BAR Tregs can suppress the response to FVIII suggests that bystander suppression can occur in the local milieu in this system. Transwell experiments confirmed that the suppression was contact-dependent. Moreover, even in the presence of antibodies to FVIII (so-called inhibitors), similarly prepared CD4+CD25 hi CD127 low A2-BAR human natural Tregs completely suppressed polyclonal anti-FVIII ASC formation. In conclusion, we demonstrated in vitro that FVIII domain-expressing BAR Tregs could efficiently target and suppress FVIII-specific memory B cells.


Asunto(s)
Linfocitos B/inmunología , Ingeniería Celular/métodos , Factor VIII/inmunología , Factor VIII/farmacología , Memoria Inmunológica/genética , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T Reguladores/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Células Cultivadas , Factor VIII/genética , Femenino , Hemofilia A/tratamiento farmacológico , Hemofilia A/inmunología , Inmunización/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Quiméricos de Antígenos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/farmacología , Transducción Genética/métodos
10.
Clin Immunol ; 207: 49-54, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31325629

RESUMEN

Allergy is a major public health concern, the main treatment for which is symptomatic relief with anti-inflammatory drugs. A key clinical challenge is to induce specific tolerance in order to control allergen-specific memory B and T cells, and specifically block effector cell responses. Our lab recently developed antigen-specific regulatory T-cell (Treg) therapies as a treatment for adverse responses. Recently, we created a chimeric antigen receptor (CAR) approach in which we engineered a target protein antigen, ovalbumin (OVA), linked with the transmembrane and signal transduction domains, CD28-CD3ζ to directly target B cells and sensitized mast cells in an allergy model. We named this receptor "BAR" for B-cell Antibody Receptor. Murine or human Tregs, transduced with a BAR containing OVA or control Tregs expressing an unrelated antigen, were successfully expanded in vitro and tested in the murine OVA-alum allergy model with measurable titers of anti-OVA IgE. Because BAR Tregs express the target antigen and could interact with specific IgE on sensitized mast cells, we first demonstrated that intravenously injected OVA-BAR Tregs did not directly lead to a drop in temperature or release of mediators in plasma indicative of anaphylaxis. Forty-eight hours later, mice were challenged intraperitoneally with 200 µg OVA to induce an anaphylactic reaction, and temperature immediately measured for 30 min. We found that OVA-BAR Tregs protected mice from hypothermia, whereas mice given control BARs (expressing an unrelated antigen) or PBS showed substantial temperature drops indicative of anaphylaxis when systemically challenged with OVA. Importantly, this effect was also demonstrated in a passive anaphylaxis model in which mice that received anti-OVA IgE antibody were protected from hypothermia when treated with OVA-BAR Tregs prior to systemic OVA challenge. These results provide proof of principle that engineered allergen-specific T-regulatory cells can provide clinical protection against severe allergic reactions in individuals already IgE-sensitized to an allergen.


Asunto(s)
Anafilaxia/prevención & control , Ovalbúmina/inmunología , Linfocitos T Reguladores/metabolismo , Alérgenos/inmunología , Animales , Femenino , Tolerancia Inmunológica/inmunología , Inmunización Pasiva , Masculino , Ratones , Ratones Endogámicos BALB C
11.
J Immunol ; 201(5): 1434-1441, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30021767

RESUMEN

Controlling immune responses in autoimmunity and to biotherapeutics is an unmet need. In hemophilia, for example, up to one third of patients receiving therapeutic factor VIII (FVIII) infusions develop neutralizing Abs termed "inhibitors." To address this problem in a mouse model of hemophilia A, we used an Ag-specific regulatory T cell (Treg) approach in which we created a novel B cell-targeting chimeric receptor composed of an FVIII Ag domain linked with the CD28-CD3ζ transmembrane and signaling domains. We termed these "BAR" for B cell-targeting Ab receptors. CD4+CD25hiCD127low human Tregs were retrovirally transduced to express a BAR containing the immunodominant FVIII C2 or A2 domains (C2- and A2-BAR). Such BAR-Tregs specifically suppressed the recall Ab response of spleen cultures from FVIII-immunized mice in vitro and completely prevented anti-FVIII Ab development in response to FVIII immunization. Mechanistic studies with purified B cells and T cells from tolerized or control recipients demonstrated that the FVIII-specific B cells were directly suppressed or anergized, whereas the T cell response remained intact. Taken together, we report in this study a successful proof-of-principle strategy using Ag-expressing Tregs to directly target specific B cells, an approach which could be adapted to address other adverse immune responses as well.


Asunto(s)
Antígenos/inmunología , Linfocitos B/inmunología , Factor VIII/inmunología , Terapia Genética , Hemofilia A/inmunología , Linfocitos T Reguladores/inmunología , Transducción Genética , Animales , Antígenos/genética , Linfocitos B/patología , Factor VIII/genética , Expresión Génica , Hemofilia A/genética , Hemofilia A/patología , Hemofilia A/terapia , Humanos , Ratones , Ratones Noqueados , Linfocitos T Reguladores/patología
12.
J Autoimmun ; 92: 77-86, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29857928

RESUMEN

Expanded polyclonal T regulatory cells (Tregs) offer great promise for the treatment of immune-mediated diseases. Inhibition by Tregs is under the control of the T-cell receptor (TCR). Therefore, we created Tregs with defined antigen specificity, using a recombinant T-cell receptor isolated from a myelin-basic protein specific T-cell clone of a multiple sclerosis (MS) patient (Ob2F3). We expressed this TCR using a retroviral expression vector in human Tregs from peripheral blood. We observed that transduced Tregs were activated in vitro in response to myelin basic protein (MBP) peptide on DR15 antigen-presenting cells (APC) and upregulated Treg markers, Foxp3, LAP and Helios. These engineered MBP-specific Tregs could suppress MBP-specific T effector cells, and were also able to suppress T cells with other specificities after Tregs had been activated through the TCR. Importantly, we showed that these engineered Tregs were able to function effectively in the presence of strong TLR-induced inflammatory signals, and that MBP-specific Tregs ameliorated EAE in myelin oligodendrocyte glycoprotein (MOG)-immunized DR15 transgenic mice. We further demonstrated in vitro that IL-2 produced by neighboring effector T cells activated MBP-specific Tregs, initiating contact-independent suppression to T effectors in local milieu. Mechanistic studies demonstrated that bystander suppression in vivo may involve transfer of soluble mediators, enhanced by cell contact between Tregs and effectors. Taken together, we show that engineered clonal MBP-specific Tregs are able to suppress autoimmune pathology in EAE. This approach may serve as a cellular therapy for MS patients with the common DR15 haplotype that is associated with disease susceptibility.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Inmunoterapia Adoptiva/métodos , Esclerosis Múltiple/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Animales , Autoinmunidad , Efecto Espectador , Células Cultivadas , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/terapia , Factores de Transcripción Forkhead/metabolismo , Ingeniería Genética , Predisposición Genética a la Enfermedad , Subtipos Serológicos HLA-DR/genética , Humanos , Interleucina-2/metabolismo , Activación de Linfocitos , Ratones , Ratones Transgénicos , Esclerosis Múltiple/genética , Esclerosis Múltiple/terapia , Proteína Básica de Mielina/inmunología , Glicoproteína Mielina-Oligodendrócito/inmunología , Polimorfismo Genético , Receptores de Antígenos de Linfocitos T/genética , Especificidad del Receptor de Antígeno de Linfocitos T
13.
Front Immunol ; 8: 1117, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28983300

RESUMEN

Human regulatory CD4+ T cells (Tregs) are potent immunosuppressive lymphocytes responsible for immune tolerance and homeostasis. Since the seminal reports identifying Tregs, vast research has been channeled into understanding their genesis, signature molecular markers, mechanisms of suppression, and role in disease. This research has opened the doors for Tregs as a potential therapeutic for diseases and disorders such as multiple sclerosis, type I diabetes, transplantation, and immune responses to protein therapeutics, like factor VIII. Seminal clinical trials have used polyclonal Tregs, but the frequency of antigen-specific Tregs among polyclonal populations is low, and polyclonal Tregs may risk non-specific immunosuppression. Antigen-specific Treg therapy, which uses genetically modified Tregs expressing receptors specific for target antigens, greatly mitigates this risk. Building on the principles of T-cell receptor cloning, chimeric antigen receptors (CARs), and a novel CAR derivative, called B-cell antibody receptors, our lab has developed different types of antigen-specific Tregs. This review discusses the current research and optimization of gene-modified antigen-specific human Tregs in our lab in several disease models. The preparations and considerations for clinical use of such Tregs also are discussed.

14.
Blood ; 129(2): 238-245, 2017 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-28064157

RESUMEN

Replacement therapy with factor VIII (FVIII) is used in patients with hemophilia A for treatment of bleeding episodes or for prophylaxis. A common and serious problem with this therapy is the patient's immune response to FVIII, because of a lack of tolerance, leading to the formation of inhibitory antibodies. Development of tolerogenic therapies, other than standard immune tolerance induction (ITI), is an unmet goal. We previously generated engineered antigen-specific regulatory T cells (Tregs), created by transduction of a recombinant T-cell receptor (TCR) isolated from a hemophilia A subject's T-cell clone. The resulting engineered T cells suppressed both T- and B-cell effector responses to FVIII. In this study, we have engineered an FVIII-specific chimeric antigen receptor (ANS8 CAR) using a FVIII-specific scFv derived from a synthetic phage display library. Transduced ANS8 CAR T cells specific for the A2 domain proliferated in response to FVIII and ANS8 CAR Tregs were able to suppress the proliferation of FVIII-specific T-effector cells with specificity for a different FVIII domain in vitro. These data suggest that engineered cells are able to promote bystander suppression. Importantly, ANS8 CAR-transduced Tregs also were able to suppress the recall antibody response of murine splenocytes from FVIII knockout mice to FVIII in vitro and in vivo. In conclusion, CAR-transduced Tregs are a promising approach for future tolerogenic treatment of hemophilia A patients with inhibitors.


Asunto(s)
Ingeniería Celular/métodos , Factor VIII/inmunología , Terapia de Inmunosupresión/métodos , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Animales , Linfocitos B , Citometría de Flujo , Humanos , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Transducción Genética
15.
Cell Immunol ; 301: 74-81, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26687613

RESUMEN

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


Asunto(s)
Factor VIII/administración & dosificación , Hemofilia A/inmunología , Tolerancia Inmunológica/inmunología , Inmunosupresores/administración & dosificación , Nanopartículas , Sirolimus/administración & dosificación , Animales , Anticuerpos Neutralizantes/inmunología , Modelos Animales de Enfermedad , Factor VIII/inmunología , Inmunosupresores/inmunología , Ratones , Sirolimus/inmunología , Vacunas Sintéticas
16.
Int J Clin Exp Med ; 8(8): 13118-26, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26550234

RESUMEN

The study was to explore diagnostic value of the virtual touch tissue imaging quantification (VTIQ) in distinguishing benign and malignant breast lesions of variable sizes. We performed conventional ultrasound and VTIQ in 139 breast lesions. The lesions were categorized into three groups according to size (group 1, ≤ 10 mm; group 2, 10-20 mm; and group 3, > 20 mm), and their mean, min, and max shear wave velocities (SWVs) were measured. Diagnoses were confirmed by pathological examination after surgery or needle biopsy. Receiver-operating characteristic curves (ROC) were constructed to determine the optimum cut-off values, calculate the area under curve (AUC), the sensitivity, specificity and accuracy for each velocity. For all groups, the mean, min, and max SWVs of malignant lesions were significantly higher than those of benign lesions (P < 0.05). The cut-off values of mean, min, and max SWVs were not significantly different among the three groups. In addition, the diagnostic performance of mean, min, and max SWV values is analogous, regardless of lesion size. In conclusion, VTIQ is a strong complement to conventional ultrasound, which is a promising method in the differential diagnosis of the breast lesions with different sizes. Further studies validate our results as well as reduce the number of unnecessary biopsies, regardless of size is warranted.

17.
PLoS One ; 10(1): e0116577, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25615825

RESUMEN

The development of neutralizing anti-factor VIII (FVIII) antibodies complicates the treatment of many hemophilia A patients. The C-terminal C2 domain is a particularly antigenic FVIII region. A crystal structure of recombinant FVIII-C2 bound to an Fab fragment of the patient-derived monoclonal antibody BO2C11, which recognizes an immunodominant inhibitor epitope on FVIII and blocks its ability to bind von Willebrand factor (VWF) and phospholipids, revealed that 15 amino acids in FVIII contact this antibody. Forty-three recombinant FVIII-C2 proteins, each with a surface-exposed side chain mutated to alanine or another residue, were generated, and surface plasmon resonance studies were carried out to evaluate effects of these substitutions on BO2C11/FVIII-C2 binding affinity. Thermodynamic analysis of experiments carried out at three temperatures indicated that one beta hairpin turn at the antigen-antibody interface (FVIII-F2196, N2198, M2199 and F2200) plus two non-contiguous arginines (FVIII-R2215 and R2220), contributed appreciably to the affinity. B-domain-deleted (BDD) FVIII-F2196A, FVIII-F2196K and FVIII-M2199A were generated and characterized. Their pro-coagulant activities and binding to VWF were similar to those of WT-BDD-FVIII, and FVIII-F2196K avoided neutralization by BO2C11 and murine inhibitory mAb 1B5. This study suggests specific sites for amino acid substitutions to rationally design FVIII variants capable of evading immunodominant neutralizing anti-FVIII antibodies.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Epítopos/química , Factor VIII/química , Factor VIII/metabolismo , Fragmentos Fab de Inmunoglobulinas/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Sitios de Unión , Factor VIII/genética , Humanos , Modelos Moleculares , Resonancia por Plasmón de Superficie , Termodinámica , Factor de von Willebrand/metabolismo
18.
Proc Natl Acad Sci U S A ; 112(2): E156-65, 2015 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-25548186

RESUMEN

Current treatments to control pathological or unwanted immune responses often use broadly immunosuppressive drugs. New approaches to induce antigen-specific immunological tolerance that control both cellular and humoral immune responses are desirable. Here we describe the use of synthetic, biodegradable nanoparticles carrying either protein or peptide antigens and a tolerogenic immunomodulator, rapamycin, to induce durable and antigen-specific immune tolerance, even in the presence of potent Toll-like receptor agonists. Treatment with tolerogenic nanoparticles results in the inhibition of CD4+ and CD8+ T-cell activation, an increase in regulatory cells, durable B-cell tolerance resistant to multiple immunogenic challenges, and the inhibition of antigen-specific hypersensitivity reactions, relapsing experimental autoimmune encephalomyelitis, and antibody responses against coagulation factor VIII in hemophilia A mice, even in animals previously sensitized to antigen. Only encapsulated rapamycin, not the free form, could induce immunological tolerance. Tolerogenic nanoparticle therapy represents a potential novel approach for the treatment of allergies, autoimmune diseases, and prevention of antidrug antibodies against biologic therapies.


Asunto(s)
Antígenos/administración & dosificación , Antígenos/química , Tolerancia Inmunológica , Terapia de Inmunosupresión/métodos , Nanopartículas/química , Animales , Linfocitos T CD4-Positivos/inmunología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/prevención & control , Encefalomielitis Autoinmune Experimental/terapia , Factor VIII/inmunología , Femenino , Hemocianinas/administración & dosificación , Hemofilia A/inmunología , Hemofilia A/terapia , Humanos , Hipersensibilidad Tardía/inmunología , Hipersensibilidad Tardía/terapia , Inmunidad Humoral , Inmunosupresores/administración & dosificación , Ácido Láctico/química , Ratones , Ratones Endogámicos BALB C , Nanocápsulas/administración & dosificación , Nanocápsulas/química , Nanopartículas/administración & dosificación , Oligodesoxirribonucleótidos/administración & dosificación , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/inmunología , Péptidos/administración & dosificación , Péptidos/química , Péptidos/inmunología , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Proteínas/administración & dosificación , Proteínas/química , Proteínas/inmunología , Proteínas Recombinantes/inmunología , Sirolimus/administración & dosificación
19.
Blood ; 125(7): 1107-15, 2015 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-25498909

RESUMEN

Expansion of human regulatory T cells (Tregs) for clinical applications offers great promise for the treatment of undesirable immune responses in autoimmunity, transplantation, allergy, and antidrug antibody responses, including inhibitor responses in hemophilia A patients. However, polyclonal Tregs are nonspecific and therefore could potentially cause global immunosuppression. To avoid this undesirable outcome, the generation of antigen-specific Tregs would be advantageous. Herein, we report the production and properties of engineered antigen-specific Tregs, created by transduction of a recombinant T-cell receptor obtained from a hemophilia A subject's T-cell clone, into expanded human FoxP3(+) Tregs. Such engineered factor VIII (FVIII)-specific Tregs efficiently suppressed the proliferation and cytokine production of FVIII-specific T-effector cells. Moreover, studies with an HLA-transgenic, FVIII-deficient mouse model demonstrated that antibody production from FVIII-primed spleen cells in vitro were profoundly inhibited in the presence of these FVIII-specific Tregs, suggesting potential utility to treat anti-FVIII inhibitory antibody formation in hemophilia A patients.


Asunto(s)
Linfocitos B Reguladores/inmunología , Ingeniería Celular , Factor VIII/inmunología , Tolerancia Inmunológica , Especificidad del Receptor de Antígeno de Linfocitos T , Linfocitos T Reguladores/inmunología , Adulto , Anciano , Animales , Linfocitos B Reguladores/metabolismo , Ingeniería Celular/métodos , Células Cultivadas , Factor VIII/genética , Factor VIII/metabolismo , Ingeniería Genética , Terapia Genética , Hemofilia A/genética , Hemofilia A/inmunología , Humanos , Tolerancia Inmunológica/genética , Masculino , Ratones , Persona de Mediana Edad , Especificidad del Receptor de Antígeno de Linfocitos T/genética , Linfocitos T Reguladores/metabolismo , Adulto Joven
20.
Chin J Cancer ; 32(3): 130-5, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22854065

RESUMEN

Despite its dual role in determining cell fate in a wide array of solid cancer cell lines, autophagy has been robustly shown to suppress or kill acute myeloid leukemia cells via degradation of the oncogenic fusion protein that drives leukemogenesis. However, autophagy also induces the demise of acute leukemia cells that do not express the known fusion protein, though the molecular mechanism remains elusive. Nevertheless, since it can induce cooperation with apoptosis and differentiation in response to autophagic signals, autophagy can be manipulated for a better therapy on acute myeloid leukemia.


Asunto(s)
Autofagia , Leucemia Mieloide Aguda/patología , Proteínas de Fusión Oncogénica/metabolismo , Antineoplásicos/uso terapéutico , Apoptosis , Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patología , Terapia Molecular Dirigida , Tretinoina/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...