Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Small ; 20(6): e2303494, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37794621

RESUMEN

Insufficient bone formation and excessive bone resorption caused by estrogen deficiency are the major factors resulting in the incidence of postmenopausal osteoporosis (PMOP). The existing drugs usually fail to re-establish the osteoblast/osteoclast balance from both sides and generate side-effects owing to the lack of bone-targeting ability. Here, engineered cell-membrane-coated nanogels PNG@mR&C capable of scavenging receptor activator of nuclear factor-κB ligand (RANKL) and responsively releasing therapeutic PTH 1-34 in the bone microenvironment are prepared from RANK and CXCR4 overexpressed bone mesenchymal stem cell (BMSC) membrane-coated chitosan biopolymers. The CXCR4 on the coated-membranes confer bone-targeting ability, and abundant RANK effectively absorb RANKL to inhibit osteoclastogenesis. Meanwhile, the release of PTH 1-34 triggered by osteoclast-mediated acid microenvironment promote osteogenesis. In addition, the dose and frequency are greatly reduced due to the smart release property, prolonged circulation time, and bone-specific accumulation. Thus, PNG@mR&C exhibits satisfactory therapeutic effects in the ovariectomized (OVX) mouse model. This study provides a new paradigm re-establishing the bone metabolic homeostasis from multitargets and shows great promise for the treatment of PMOP.


Asunto(s)
Osteoclastos , Osteoporosis Posmenopáusica , Humanos , Animales , Ratones , Femenino , Osteoporosis Posmenopáusica/tratamiento farmacológico , Osteoporosis Posmenopáusica/metabolismo , Nanogeles , Biomimética , Diferenciación Celular , Osteoblastos , Osteogénesis , FN-kappa B/metabolismo
2.
J Mater Chem B ; 11(13): 2946-2957, 2023 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-36916173

RESUMEN

Endogenous repair of critical bone defects is typically hampered by inadequate vascularization in the early stages and insufficient bone regeneration later on. Therefore, drug delivery systems with the ability to couple angiogenesis and osteogenesis in a spatiotemporal manner are highly desirable for vascularized bone formation. Herein, we devoted to develop a liquid crystal formulation system (LCFS) attaining a controlled temporal release of angiogenic and osteoinductive bioactive molecules that could orchestrate the coupling of angiogenesis and osteogenesis in an optimal way. It has been demonstrated that the release kinetics of biomolecules depend on the hydrophobicity of the loaded molecules, making the delivery profile programmable and controllable. The hydrophilic deferoxamine (DFO) could be released rapidly within 5 days to activate angiogenic signaling, while the lipophilic simvastatin (SIM) showed a slow and sustained release for continuous osteogenic induction. Apart from its good biocompatibility with mesenchymal stem cells derived from rat bone marrow (rBMSCs), the DFO/SIM loaded LCFS could stimulate the formation of a vascular morphology in human umbilical vein endothelial cells (HUVECs) and the osteogenic differentiation of rBMSCs in vitro. The in vivo rat femoral defect models have witnessed the prominent angiogenic and osteogenic effects induced by the sequential presentation of DFO and SIM. This study suggests that the sequential release of DFO and SIM from the LCFS results in enhanced bone formation, offering a facile and viable treatment option for bone defects by mimicking the physiological process of bone regeneration.


Asunto(s)
Cristales Líquidos , Osteogénesis , Ratas , Humanos , Animales , Ratas Sprague-Dawley , Regeneración Ósea , Células Endoteliales de la Vena Umbilical Humana
3.
ACS Nano ; 16(7): 11076-11091, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35801837

RESUMEN

Recently, bone marrow endothelial cells (BMECs) were found to play an important role in regulating bone homeostasis. However, few studies utilized BMECs to treat bone metabolic diseases including osteoporosis. Here, we reported bioinspired nanovesicles (BNVs) prepared from human induced pluripotent stem cells-derived endothelial cells under hypoxia culture through an extrusion approach. Abundant membrane C-X-C motif chemokine receptor 4 conferred these BNVs bone-targeting ability and the endothelial homology facilitated the BMEC tropism. Due to their unique endogenous miRNA cargos, these BNVs re-educated BMECs to secret cytokines favoring osteogenesis and anti-inflammation. Owing to the conversion of secretory phenotype, the osteogenic differentiation of bone mesenchymal stem cells was facilitated, and the M1-macrophage-dominant pro-inflammatory microenvironment was ameliorated in osteoporotic bones. Taken together, this study proposed BMEC-targeting nanovesicles treating osteoporosis via converting the skeletal endothelium-associated secretory phenotype.


Asunto(s)
Células Madre Pluripotentes Inducidas , Osteoporosis , Humanos , Osteogénesis , Células Endoteliales/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Osteoporosis/tratamiento farmacológico , Diferenciación Celular/fisiología , Endotelio/metabolismo , Fenotipo , Células Cultivadas
4.
Appl Bionics Biomech ; 2022: 7194855, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35450147

RESUMEN

To evaluate the protection performance of SPD (surge protective device) against electromagnetic pulse, the response ability of several typical surge protective devices to wide and narrow electromagnetic pulses was tested by using a SPD response ability test system. The results showed that SPD commonly used in lightning surge protection had certain ability to suppress electromagnetic pulse conduction disturbance. Gas discharge tubes presented typical clamping characteristics for wide pulses. MOV and TVS had obvious clamping effect on wide pulses, while had no clamping effect on narrow pulses, but could obviously reduce its peak value. Zener diodes had obvious clamping effect on narrow pulses, and the clamping voltage control accuracy was high.

5.
Bioact Mater ; 10: 207-221, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34901540

RESUMEN

The complex pathogenesis of osteoporosis includes excessive bone resorption, insufficient bone formation and inadequate vascularization, a combination which is difficult to completely address with conventional therapies. Engineered exosomes carrying curative molecules show promise as alternative osteoporosis therapies, but depend on specifically-functionalized vesicles and appropriate engineering strategies. Here, we developed an exosome delivery system based on exosomes secreted by mesenchymal stem cells (MSCs) derived from human induced pluripotent stem cells (iPSCs). The engineered exosomes BT-Exo-siShn3, took advantage of the intrinsic anti-osteoporosis function of these special MSC-derived exosomes and collaborated with the loaded siRNA of the Shn3 gene to enhance the therapeutic effects. Modification of a bone-targeting peptide endowed the BT-Exo-siShn3 an ability to deliver siRNA to osteoblasts specifically. Silencing of the osteoblastic Shn3 gene enhanced osteogenic differentiation, decreased autologous RANKL expression and thereby inhibited osteoclast formation. Furthermore, Shn3 gene silencing increased production of SLIT3 and consequently facilitated vascularization, especially formation of type H vessels. Our study demonstrated that BT-Exo-siShn3 could serve as a promising therapy to kill three birds with one stone and implement comprehensive anti-osteoporosis effects.

6.
J Nanobiotechnology ; 19(1): 248, 2021 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-34407835

RESUMEN

BACKGROUND: Osteoporosis (OP) is a systemic skeletal disease marked by bone mass reduction and bone tissue destruction. Hormone replacement therapy is an effective treatment for post-menopausal OP, but estrogen has poor tissue selectivity and severe side effects. RESULTS: In this study, we constructed a poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs)-based drug delivery system to co-load 17ß estradiol (E2) and iron oxide (Fe3O4) together, modified with alendronate (AL) to achieve bone targeting and realize a magnetically remote-controllable drug release. The NPs were fabricated through the emulsion solvent diffusion method. The particle size was approximately 200 nm while the encapsulation efficiency of E2 was 58.34 ± 9.21%. The NPs were found to be spherical with a homogenous distribution of particle size. The NPs showed good stability, good biocompatibility, high encapsulation ability of E2 and excellent magnetic properties. The NPs could be effectively taken up by Raw 264.7 cells and were effective in enriching drugs in bone tissue. The co-loaded NPs exposed to an external magnetic field ameliorated OVX-induced bone loss through increased BV/TV, decreased Tb.N and Tb.Sp, improved bone strength, increased PINP and OC, and downregulated CTX and TRAP-5b. The haematological index and histopathological analyses displayed the NPs had less side effects on non-skeletal tissues. CONCLUSIONS: This study presented a remote-controlled release system based on bone-targeted multifunctional NPs and a new potential approach to bone-targeted therapy of OP.


Asunto(s)
Huesos/efectos de los fármacos , Estradiol/farmacología , Osteoporosis/tratamiento farmacológico , Ovariectomía/efectos adversos , Animales , Sistemas de Liberación de Medicamentos/métodos , Liberación de Fármacos , Emulsiones/uso terapéutico , Estradiol/química , Femenino , Compuestos Férricos , Ratones , Nanopartículas , Tamaño de la Partícula , Células RAW 264.7 , Ratas , Ratas Sprague-Dawley , Fosfatasa Ácida Tartratorresistente
7.
Redox Biol ; 41: 101915, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33662874

RESUMEN

Senile osteoporosis (SOP) is widely regarded as one of the typical aging-related diseases due to a decrease in bone mass and the destruction in microarchitecture. The inhibition of mitophagy can promote bone marrow mesenchymal stem cells (BMSCs) senescence, and increasing studies have shown that interventions targeting BMSCs senescence can ameliorate osteoporosis, exhibiting their potential for use as therapeutic strategies. Sirtuin-3 (Sirt3) is an essential mitochondria metabolic regulatory enzyme that plays an important role in mitochondrial homeostasis, but its role in bone homeostasis remains largely unknown. This study seeks to investigate whether advanced glycation end products (AGEs) accumulation aggravated BMSCs senescence and SOP, and explored the mechanisms underlying these effects. We observed that AGEs significantly aggravated BMSCs senescence, as well as promoted mitochondrial dysfunction and inhibited mitophagy in a concentration-dependent manner. In addition, this effect could be further strengthened by Sirt3 silencing. Importantly, we identified that the reduction of Sirt3 expression and the mitophagy were vital mechanisms in AGEs-induced BMSCs senescence. Furthermore, overexpression of Sirt3 by intravenously injection with recombinant adeno-associated virus 9 carrying Sirt3 plasmids (rAAV-Sirt3) significantly alleviated BMSCs senescence and the formation of SOP in SAMP6. In conclusion, our data demonstrated that Sirt3 protects against AGEs-induced BMSCs senescence and SOP. Targeting Sirt3 to improve mitophagy may represent a potential therapeutic strategy for attenuating AGEs-associated SOP.


Asunto(s)
Células Madre Mesenquimatosas , Osteoporosis , Sirtuina 3 , Envejecimiento , Senescencia Celular , Humanos , Mitofagia
8.
Clin Cancer Res ; 25(8): 2560-2574, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30655315

RESUMEN

PURPOSE: Patients with relapsed pediatric solid tumors and CNS malignancies have few therapeutic options and frequently die of their disease. Chimeric antigen receptor (CAR) T cells have shown tremendous success in treating relapsed pediatric acute lymphoblastic leukemia, but this has not yet translated to treating solid tumors. This is partially due to a paucity of differentially expressed cell surface molecules on solid tumors that can be safely targeted. Here, we present B7-H3 (CD276) as a putative target for CAR T-cell therapy of pediatric solid tumors, including those arising in the central nervous system. EXPERIMENTAL DESIGN: We developed a novel B7-H3 CAR whose binder is derived from a mAb that has been shown to preferentially bind tumor tissues and has been safely used in humans in early-phase clinical trials. We tested B7-H3 CAR T cells in a variety of pediatric cancer models. RESULTS: B7-H3 CAR T cells mediate significant antitumor activity in vivo, causing regression of established solid tumors in xenograft models including osteosarcoma, medulloblastoma, and Ewing sarcoma. We demonstrate that B7-H3 CAR T-cell efficacy is largely dependent upon high surface target antigen density on tumor tissues and that activity is greatly diminished against target cells that express low levels of antigen, thus providing a possible therapeutic window despite low-level normal tissue expression of B7-H3. CONCLUSIONS: B7-H3 CAR T cells could represent an exciting therapeutic option for patients with certain lethal relapsed or refractory pediatric malignancies, and should be tested in carefully designed clinical trials.


Asunto(s)
Antígenos de Neoplasias/inmunología , Antígenos B7/inmunología , Neoplasias Encefálicas/etiología , Neoplasias Encefálicas/metabolismo , Inmunoterapia Adoptiva , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Antígenos B7/antagonistas & inhibidores , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Inmunohistoquímica , Inmunoterapia Adoptiva/métodos , Ratones , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Front Immunol ; 8: 1899, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29354121

RESUMEN

NEO-201 is a novel humanized IgG1 monoclonal antibody that was derived from an immunogenic preparation of tumor-associated antigens from pooled allogeneic colon tumor tissue extracts. It was found to react against a variety of cultured human carcinoma cell lines and was highly reactive against the majority of tumor tissues from many different carcinomas, including colon, pancreatic, stomach, lung, and breast cancers. NEO-201 also exhibited tumor specificity, as the majority of normal tissues were not recognized by this antibody. Functional assays revealed that treatment with NEO-201 is capable of mediating both antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) against tumor cells. Furthermore, the growth of human pancreatic xenograft tumors in vivo was largely attenuated by treatment with NEO-201 both alone and in combination with human peripheral blood mononuclear cells as an effector cell source for ADCC. In vivo biodistribution studies in human tumor xenograft-bearing mice revealed that NEO-201 preferentially accumulates in the tumor but not organ tissue. Finally, a single-dose toxicity study in non-human primates demonstrated safety and tolerability of NEO-201, as a transient decrease in circulating neutrophils was the only related adverse effect observed. These findings indicate that NEO-201 warrants clinical testing as both a novel diagnostic and therapeutic agent for the treatment of a broad variety of carcinomas.

10.
Cancer Immunol Res ; 4(10): 869-880, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27549124

RESUMEN

Genetically engineered T cells expressing CD19-specific chimeric antigen receptors (CAR) have shown impressive activity against B-cell malignancies, and preliminary results suggest that T cells expressing a first-generation disialoganglioside (GD2)-specific CAR can also provide clinical benefit in patients with neuroblastoma. We sought to assess the potential of GD2-CAR therapies to treat pediatric sarcomas. We observed that 18 of 18 (100%) of osteosarcomas, 2 of 15 (13%) of rhabdomyosarcomas, and 7 of 35 (20%) of Ewing sarcomas expressed GD2. T cells engineered to express a third-generation GD2-CAR incorporating the 14g2a-scFv with the CD28, OX40, and CD3ζ signaling domains (14g2a.CD28.OX40.ζ) mediated efficient and comparable lysis of both GD2+ sarcoma and neuroblastoma cell lines in vitro However, in xenograft models, GD2-CAR T cells had no antitumor effect against GD2+ sarcoma, despite effectively controlling GD2+ neuroblastoma. We observed that pediatric sarcoma xenografts, but not neuroblastoma xenografts, induced large populations of monocytic and granulocytic murine myeloid-derived suppressor cells (MDSC) that inhibited human CAR T-cell responses in vitro Treatment of sarcoma-bearing mice with all-trans retinoic acid (ATRA) largely eradicated monocytic MDSCs and diminished the suppressive capacity of granulocytic MDSCs. Combined therapy using GD2-CAR T cells plus ATRA significantly improved antitumor efficacy against sarcoma xenografts. We conclude that retinoids provide a clinically accessible class of agents capable of diminishing the suppressive effects of MDSCs, and that co-administration of retinoids may enhance the efficacy of CAR therapies targeting solid tumors. Cancer Immunol Res; 4(10); 869-80. ©2016 AACR.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Células Supresoras de Origen Mieloide/efectos de los fármacos , Receptores de Antígenos de Linfocitos T/metabolismo , Sarcoma/terapia , Tretinoina/farmacología , Animales , Línea Celular Tumoral , Niño , Terapia Combinada , Gangliósidos/metabolismo , Humanos , Ratones Endogámicos NOD , Células Supresoras de Origen Mieloide/inmunología , Neuroblastoma/inmunología , Neuroblastoma/metabolismo , Neuroblastoma/patología , Sarcoma/inmunología , Sarcoma/metabolismo , Sarcoma/patología , Linfocitos T/inmunología , Linfocitos T/trasplante , Resultado del Tratamiento , Tretinoina/uso terapéutico , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Blood ; 125(19): 2958-67, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25814528

RESUMEN

T-cell receptors (TCRs) and chimeric antigen receptors recognizing tumor-associated antigens (TAAs) can now be engineered to be expressed on a wide array of immune effectors. Engineered receptors targeting TAAs have most commonly been expressed on mature T cells, however, some have postulated that receptor expression on immune progenitors could yield T cells with enhanced potency. We generated mice (survivin-TCR-transgenic [Sur-TCR-Tg]) expressing a TCR recognizing the immunodominant epitope (Sur20-28) of murine survivin during early stages of thymopoiesis. Spontaneous T-cell acute lymphoblastic leukemia (T-ALL) occurred in 100% of Sur-TCR-Tg mice derived from 3 separate founders. The leukemias expressed the Sur-TCR and signaled in response to the Sur20-28 peptide. In preleukemic mice, we observed increased cycling of double-negative thymocytes expressing the Sur-TCR and increased nuclear translocation of nuclear factor of activated T cells, consistent with TCR signaling induced by survivin expression in the murine thymus. ß2M(-/-) Sur-TCR-Tg mice, which cannot effectively present survivin peptides on class I major histocompatibility complex, had significantly diminished rates of leukemia. We conclude that TCR signaling during the early stages of thymopoiesis mediates an oncogenic signal, and therefore expression of signaling receptors on developing thymocytes with specificity for TAAs expressed in the thymus could pose a risk for neoplasia, independent of insertional mutagenesis.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Moléculas de Adhesión Celular/metabolismo , Transformación Celular Neoplásica , Proteínas Inhibidoras de la Apoptosis/fisiología , Proteínas de Neoplasias/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/etiología , Receptores de Antígenos de Linfocitos T/fisiología , Proteínas Represoras/fisiología , Subgrupos de Linfocitos T/inmunología , Timo/inmunología , Animales , Antígenos de Neoplasias/genética , Western Blotting , Moléculas de Adhesión Celular/genética , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Proteínas de Homeodominio/fisiología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Neoplasias/genética , Fragmentos de Péptidos/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Survivin , Timo/citología , Timo/metabolismo , Células Tumorales Cultivadas
12.
Lancet ; 385(9967): 517-528, 2015 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-25319501

RESUMEN

BACKGROUND: Chimeric antigen receptor (CAR) modified T cells targeting CD19 have shown activity in case series of patients with acute and chronic lymphocytic leukaemia and B-cell lymphomas, but feasibility, toxicity, and response rates of consecutively enrolled patients treated with a consistent regimen and assessed on an intention-to-treat basis have not been reported. We aimed to define feasibility, toxicity, maximum tolerated dose, response rate, and biological correlates of response in children and young adults with refractory B-cell malignancies treated with CD19-CAR T cells. METHODS: This phase 1, dose-escalation trial consecutively enrolled children and young adults (aged 1-30 years) with relapsed or refractory acute lymphoblastic leukaemia or non-Hodgkin lymphoma. Autologous T cells were engineered via an 11-day manufacturing process to express a CD19-CAR incorporating an anti-CD19 single-chain variable fragment plus TCR zeta and CD28 signalling domains. All patients received fludarabine and cyclophosphamide before a single infusion of CD19-CAR T cells. Using a standard 3 + 3 design to establish the maximum tolerated dose, patients received either 1 × 10(6) CAR-transduced T cells per kg (dose 1), 3 × 10(6) CAR-transduced T cells per kg (dose 2), or the entire CAR T-cell product if sufficient numbers of cells to meet the assigned dose were not generated. After the dose-escalation phase, an expansion cohort was treated at the maximum tolerated dose. The trial is registered with ClinicalTrials.gov, number NCT01593696. FINDINGS: Between July 2, 2012, and June 20, 2014, 21 patients (including eight who had previously undergone allogeneic haematopoietic stem-cell transplantation) were enrolled and infused with CD19-CAR T cells. 19 received the prescribed dose of CD19-CAR T cells, whereas the assigned dose concentration could not be generated for two patients (90% feasible). All patients enrolled were assessed for response. The maximum tolerated dose was defined as 1 × 10(6) CD19-CAR T cells per kg. All toxicities were fully reversible, with the most severe being grade 4 cytokine release syndrome that occurred in three (14%) of 21 patients (95% CI 3·0-36·3). The most common non-haematological grade 3 adverse events were fever (nine [43%] of 21 patients), hypokalaemia (nine [43%] of 21 patients), fever and neutropenia (eight [38%] of 21 patients), and cytokine release syndrome (three [14%) of 21 patients). INTERPRETATION: CD19-CAR T cell therapy is feasible, safe, and mediates potent anti-leukaemic activity in children and young adults with chemotherapy-resistant B-precursor acute lymphoblastic leukaemia. All toxicities were reversible and prolonged B-cell aplasia did not occur. FUNDING: National Institutes of Health Intramural funds and St Baldrick's Foundation.


Asunto(s)
Antígenos CD19 , Tratamiento Basado en Trasplante de Células y Tejidos , Linfoma no Hodgkin/terapia , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Receptores de Antígenos de Linfocitos T , Linfocitos T/trasplante , Adolescente , Adulto , Niño , Preescolar , Quimera , Estudios de Cohortes , Estudios de Factibilidad , Femenino , Humanos , Lactante , Masculino , Linfocitos T/inmunología , Resultado del Tratamiento , Adulto Joven
13.
Sci Transl Med ; 6(237): 237ra67, 2014 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-24848257

RESUMEN

Suppression of the host's immune system plays a major role in cancer progression. Tumor signaling of programmed death 1 (PD1) on T cells and expansion of myeloid-derived suppressor cells (MDSCs) are major mechanisms of tumor immune escape. We sought to target these pathways in rhabdomyosarcoma (RMS), the most common soft tissue sarcoma of childhood. Murine RMS showed high surface expression of PD-L1, and anti-PD1 prevented tumor growth if initiated early after tumor inoculation; however, delayed anti-PD1 had limited benefit. RMS induced robust expansion of CXCR2(+)CD11b(+)Ly6G(hi) MDSCs, and CXCR2 deficiency prevented CD11b(+)Ly6G(hi) MDSC trafficking to the tumor. When tumor trafficking of MDSCs was inhibited by CXCR2 deficiency, or after anti-CXCR2 monoclonal antibody therapy, delayed anti-PD1 treatment induced significant antitumor effects. Thus, CXCR2(+)CD11b(+)Ly6G(hi) MDSCs mediate local immunosuppression, which limits the efficacy of checkpoint blockade in murine RMS. Human pediatric sarcomas also produce CXCR2 ligands, including CXCL8. Patients with metastatic pediatric sarcomas display elevated serum CXCR2 ligands, and elevated CXCL8 is associated with diminished survival in this population. We conclude that accumulation of MDSCs in the tumor bed limits the efficacy of checkpoint blockade in cancer. We also identify CXCR2 as a novel target for modulating tumor immune escape and present evidence that CXCR2(+)CD11b(+)Ly6G(hi) MDSCs are an important suppressive myeloid subset in pediatric sarcomas. These findings present a translatable strategy to improve the efficacy of checkpoint blockade by preventing trafficking of MDSCs to the tumor site.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Quimiotaxis/efectos de los fármacos , Células Madre Neoplásicas/efectos de los fármacos , Receptores de Interleucina-8B/antagonistas & inhibidores , Rabdomiosarcoma/tratamiento farmacológico , Escape del Tumor/efectos de los fármacos , Adolescente , Adulto , Animales , Anticuerpos Monoclonales/administración & dosificación , Antígenos Ly/metabolismo , Antígeno B7-H1/metabolismo , Antígeno CD11b/metabolismo , Estudios de Casos y Controles , Línea Celular Tumoral , Quimiocina CXCL1/sangre , Niño , Preescolar , Citocinas/metabolismo , Femenino , Humanos , Interleucina-8/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Pronóstico , Receptores de Interleucina-8B/deficiencia , Receptores de Interleucina-8B/genética , Rabdomiosarcoma/sangre , Rabdomiosarcoma/inmunología , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/patología , Factores de Tiempo , Carga Tumoral/efectos de los fármacos , Microambiente Tumoral , Adulto Joven
14.
PLoS One ; 8(3): e58860, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23554941

RESUMEN

Although tumors naturally prime adaptive immune responses, tolerance may limit the capacity to control progression and can compromise effectiveness of immune-based therapies for cancer. Post-proline cleaving enzymes (PPCE) modulate protein function through N-terminal dipeptide cleavage and inhibition of these enzymes has been shown to have anti-tumor activity. We investigated the mechanism by which Val-boroPro, a boronic dipeptide that inhibits post-proline cleaving enzymes, mediates tumor regression and tested whether this agent could serve as a novel immune adjuvant to dendritic cell vaccines in two different murine syngeneic murine tumors. In mice challenged with MB49, which expresses the HY antigen complex, T cell responses primed by the tumor with and without Val-boroPro were measured using interferon gamma ELISPOT. Antibody depletion and gene-deficient mice were used to establish the immune cell subsets required for tumor regression. We demonstrate that Val-boroPro mediates tumor eradication by accelerating the expansion of tumor-specific T cells. Interestingly, T cells primed by tumor during Val-boroPro treatment demonstrate increased capacity to reject tumors following adoptive transfer without further treatment of the recipient. Val-boroPro -mediated tumor regression requires dendritic cells and is associated with enhanced trafficking of dendritic cells to tumor draining lymph nodes. Finally, dendritic cell vaccination combined with Val-boroPro treatment results in complete regression of established tumors. Our findings demonstrate that Val-boroPro has antitumor activity and a novel mechanism of action that involves more robust DC trafficking with earlier priming of T cells. Finally, we show that Val-boroPro has potent adjuvant properties resulting in an effective therapeutic vaccine.


Asunto(s)
Ácidos Borónicos/inmunología , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Dipéptidos/inmunología , Neoplasias/inmunología , Linfocitos T/inmunología , Adyuvantes Inmunológicos , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/uso terapéutico , Línea Celular Tumoral , Quimiocinas/inmunología , Citocinas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Inmunoterapia Adoptiva , Ganglios Linfáticos/inmunología , Masculino , Ratones , Neoplasias/patología , Neoplasias/terapia , Inducción de Remisión
15.
J Immunother ; 34(2): 187-95, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21304401

RESUMEN

Natural killer (NK) cells can mediate potent antitumor effects, but factors regulating the efficiency of tumor lysis remain unclear. Studies in allogeneic stem cell transplantation highlight an important role for killer cell immunoglobulin-like receptor (KIR) mismatch in overcoming human leukocyte antigen-mediated inhibitory signals. However, other activating and inhibitory signals also modulate tumor lysis by NK cells. We used rhIL15 and artificial antigen presenting cells expressing CD137L and IL15Rα to activate and expand peripheral blood NK cells (CD137L/IL15 NK) up to 1000-fold in 3 weeks. Compared with resting NK cells, CD137L/IL15 NK cells show modest increases in KIR expression and substantial increases in NKG2D, tumor necrosis factor-related apoptosis-inducing ligand, and natural cytotoxicity receptors (NCRs: NKp30, NKp44, NKp46). Compared with resting NK cells, CD137L/IL15 NK cells mediate enhanced cytotoxicity against allogeneic and autologous tumors and KIR signaling did not substantially inhibit cytotoxicity. Rather, tumor lysis by CD137L/IL15 activated NK cells was predominantly driven by NCR signaling as blockade of NCRs dramatically diminished the lysis of a wide array of tumor targets. Furthermore, tumor lysis by CD137L/IL15 NK cells was tightly linked to NCR expression levels that peaked on day 8 to 10 after NK activation, and cytotoxicity diminished on subsequent days as NCR expression declined. We conclude that KIR mismatch is not a prerequisite for tumor killing by CD137L/IL15 NK cells and that NCR expression provides a biomarker for predicting potency of CD137L/IL15 NK cells in studies of NK cell-based immunotherapy.


Asunto(s)
Ligando 4-1BB/inmunología , Interleucina-15/inmunología , Células Asesinas Naturales/inmunología , Proteínas Recombinantes/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Línea Celular Tumoral , Proliferación Celular , Citotoxicidad Inmunológica , Humanos , Subunidad alfa del Receptor de Interleucina-15/inmunología , Células K562 , Ratones , Receptores KIR/inmunología , Receptores Gatillantes de la Citotoxidad Natural/inmunología , Transducción de Señal/inmunología
16.
Nat Immunol ; 11(3): 257-64, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20118929

RESUMEN

Immature CD4(+)CD8(+) (double-positive (DP)) thymocytes are signaled via T cell antigen receptors (TCRs) to undergo positive selection and become responsive to intrathymic cytokines such as interleukin 7 (IL-7). We report here that cytokine signaling is required for positively selected thymocytes to express the transcription factor Runx3, specify CD8 lineage choice and differentiate into cytotoxic-lineage T cells. In DP thymocytes genetically engineered to be cytokine responsive, IL-7 signaling induced TCR-unsignaled DP thymocytes to express Runx3 and to differentiate into mature CD8(+) T cells, completely circumventing positive selection. We conclude that TCR-mediated positive selection converts DP cells into cytokine-responsive thymocytes, but it is subsequent signaling by intrathymic cytokines that specifies CD8 lineage choice and promotes differentiation into cytotoxic-lineage T cells.


Asunto(s)
Citocinas/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Recuento de Células , Diferenciación Celular/inmunología , Linaje de la Célula , Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Citometría de Flujo , Interleucina-7/inmunología , Ratones , Ratones Noqueados , Ratones Transgénicos , Factor de Transcripción STAT5/inmunología , Transducción de Señal
17.
Am J Chin Med ; 38(1): 65-73, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20128045

RESUMEN

The present study investigated the effects of a flavonoid extract from Cynomorium songaricum on the swimming endurance of rats by measuring changes of free radical scavenging enzymes, such as CuZn-SOD (copper, zinc-superoxide dismutase) and GSH-px (glutathione peroxidase), and body weights. Significant and dose-dependent antioxidant and anti-fatigue effects of flavonoids (rutin, catechin and isoquercitrin) on swimming rats were observed during 10 days of swimming exercise. After treatment with the flavonoid extract at doses of 0.5, 1.0, and 2.0 g/kg body weight, the CuZn-SOD and GSH-px activities in swimming rats were increased by 1.4%, 3.3%, 4.1% and 112.2%, 208.7%, 261.7%, respectively, while the levels of MDA (malondialdehyde) were decreased by 64.7%, 79.4%, and 86.4% respectively. Furthermore, the average body weight and the total swimming time were increased by 3.1%, 8.8%, 10.6%, and 7.7%, 34.5%, 61.5%, respectively. Our experimental results suggest that flavonoid supplementation could not only reduce free radical formation and scavenge free radicals, but also enhance endurance exercise performance by reducing muscle fatigue.


Asunto(s)
Antioxidantes/farmacología , Cynomorium/química , Flavonoides/farmacología , Fatiga Muscular/efectos de los fármacos , Condicionamiento Físico Animal/fisiología , Resistencia Física/efectos de los fármacos , Extractos Vegetales/farmacología , Animales , Antioxidantes/metabolismo , Peso Corporal/efectos de los fármacos , Catequina/farmacología , Relación Dosis-Respuesta a Droga , Glutatión Peroxidasa/sangre , Masculino , Resistencia Física/fisiología , Tallos de la Planta , Quercetina/análogos & derivados , Quercetina/farmacología , Ratas , Ratas Wistar , Rutina/farmacología , Superóxido Dismutasa/sangre , Natación
18.
Blood ; 114(18): 3831-40, 2009 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-19704119

RESUMEN

Lymphopenia enhances the effectiveness of adoptive immunotherapy by facilitating expansion of transferred T cells but also limits the T-cell repertoire available to mediate immune responses and, in humans, is associated with chronic immune dysfunction. Previous studies concluded that lymphopenia augments adoptive immunotherapy by diminishing Tregs and increasing homeostatic cytokines. We sought to determine whether targeted therapies that replicate the physiology of lymphopenia in lymphoreplete hosts could provide a similarly supportive milieu. Pmel-1 T cells were transferred to B16-bearing lymphopenic versus lymphoreplete mice receiving alphaCD25 and/or recombinant human interleukin-7. Although CD25-based Treg depletion was inefficient because of peripheral expansion of CD4+CD25-FOXP3+ cells, outcomes were better in alphaCD25-treated lymphoreplete hosts than in lymphopenic hosts, and adoptive immunotherapy was most effective in lymphoreplete hosts receiving alphaCD25 plus recombinant human interleukin-7. Lymphopenic hosts supported increased proliferation of adoptively transferred antigen-specific T cells, but cells transferred to lymphoreplete recipients receiving targeted therapies showed superior function. Further, determinant spreading was substantial in lymphoreplete hosts but absent in lymphopenic hosts. These results demonstrate that targeted therapies delivered to mimic the "physiology of lymphopenia" enhance the efficacy of adoptive immunotherapy in lymphoreplete hosts and provide a potentially superior alternative to the induction of lymphopenia.


Asunto(s)
Traslado Adoptivo , Linfopenia/terapia , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/trasplante , Animales , Proliferación Celular/efectos de los fármacos , Humanos , Subunidad alfa del Receptor de Interleucina-2/inmunología , Interleucina-7/inmunología , Interleucina-7/farmacología , Linfopenia/inmunología , Linfopenia/fisiopatología , Ratones , Ratones Noqueados
19.
Nat Immunol ; 10(2): 149-57, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19136960

RESUMEN

Interleukin 7 (IL-7) and T cell antigen receptor signals have been proposed to be the main drivers of homeostatic T cell proliferation. However, it is not known why CD4(+) T cells undergo less-efficient homeostatic proliferation than CD8(+) T cells do. Here we show that systemic IL-7 concentrations increased during lymphopenia because of diminished use of IL-7 but that IL-7 signaling on IL-7 receptor-alpha-positive (IL-7Ralpha(+)) dendritic cells (DCs) in lymphopenic settings paradoxically diminished the homeostatic proliferation of CD4(+) T cells. This effect was mediated at least in part by IL-7-mediated downregulation of the expression of major histocompatibility complex class II on IL-7Ralpha(+) DCs. Our results indicate that IL-7Ralpha(+) DCs are regulators of the peripheral CD4(+) T cell niche and that IL-7 signals in DCs prevent uncontrolled CD4(+) T cell population expansion in vivo.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Células Dendríticas/metabolismo , Homeostasis/inmunología , Interleucina-7/metabolismo , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Ensayo de Inmunoadsorción Enzimática , Retroalimentación Fisiológica , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Interleucina-7/genética , Interleucina-7/inmunología , Activación de Linfocitos/inmunología , Linfopenia/inmunología , Linfopenia/metabolismo , Ratones , Ratones Mutantes , Receptores de Interleucina-7/inmunología , Receptores de Interleucina-7/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología , Células del Estroma/inmunología , Células del Estroma/metabolismo
20.
Nat Immunol ; 9(11): 1288-96, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18820682

RESUMEN

T helper type 2 (T(H)2) cells are essential for humoral immunity and host defense. Interleukin 4 (IL-4) drives T(H)2 differentiation and IL-2 augments the accessibility of Il4 chromatin. Here we demonstrate that IL-2, by inducing binding of STAT5 to the Il4ra locus, which encodes IL-4 receptor alpha-chain (IL-4Ralpha), was essential for inducing and maintaining IL-4Ralpha expression. Although IL-4 induced IL-4Ralpha expression, T cell receptor-induced IL-4Ralpha expression was normal in Il4(-/-) cells but was much lower in Il2(-/-) cells. Notably, forced IL-4Ralpha expression restored the T(H)2 differentiation of Il2(-/-) cells. Moreover, genome-wide mapping by chromatin immunoprecipitation coupled with sequencing showed broad interaction of the transcription factors STAT5A and STAT5B with genes associated with T(H)2 differentiation. Our results identify a previously unappreciated function for IL-2 in 'priming' T cells for T(H)2 differentiation and in maintaining the expression of Il4ra and other genes in T(H)2-committed cells.


Asunto(s)
Regulación de la Expresión Génica , Interleucina-2/metabolismo , Subunidad alfa del Receptor de Interleucina-4/genética , Células Th2/inmunología , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Interleucina-2/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor de Transcripción STAT5/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA