Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Curr Drug Targets ; 23(6): 597-605, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34254912

RESUMEN

Fatty acid binding protein A (A-FABP) is one of FABPs isoforms found mainly in adipose tissue and macrophages. It works through many integrated pathways, regulating inflammation and lipid metabolism, promoting glucose production, impairing insulin function, and contributing to diseases such as atherosclerosis and diabetes. A-FABP is upregulated in the adipose tissue of obese patients and its increased release into the bloodstream is positively associated with body mass index. Consequently, A-FABP plays a key role in regulating metabolism in obese people. Recent studies in mouse models and humans demonstrated the role of A-FABP in increasing the risk of obesity-related cancers. Here we summarized the state of research on the link between obesity, cancer and A-FABP as a new potential therapeutic target for the treatment of obesity - associated cancers.


Asunto(s)
Neoplasias , Obesidad , Adipocitos/metabolismo , Tejido Adiposo , Animales , Proteínas de Unión a Ácidos Grasos/metabolismo , Proteínas de Unión a Ácidos Grasos/uso terapéutico , Humanos , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Obesidad/complicaciones , Obesidad/tratamiento farmacológico , Obesidad/metabolismo
2.
Molecules ; 26(10)2021 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-34063466

RESUMEN

INTRODUCTION: Adiponectin is a hormone secreted by adipocytes, which exhibits insulin-sensitizing and anti-inflammatory properties and acts through adiponectin receptors: AdipoR1 and AdipoR2. The aim of the study was to evaluate whether activation of adiponectin receptors AdipoR1 and AdipoR2 with an orally active agonist AdipoRon has gastroprotective effect and to investigate the possible underlying mechanism. METHODS: We used two well-established mouse models of gastric ulcer (GU) induced by oral administration of EtOH (80% solution in water) or diclofenac (30 mg/kg, p.o.). Gastroprotective effect of AdipoRon (dose 5 and 50 mg /kg p.o) was compared to omeprazole (20 mg/kg p.o.) or 5% DMSO solution (control). Clinical parameters of gastroprotection were assessed using macroscopic (gastric lesion area) and microscopic (evaluation of the gastric mucosa damage) scoring. To establish the molecular mechanism, we measured: myeloperoxidase (MPO), superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPX) activities; glutathione (GSH) level; and IL-1ß, adenosine monophosphate-activated protein kinase (AMPK), and phosphorylated AMPK expression in gastric tissue. RESULTS: AdipoRon produced a gastroprotective effect in both GU mouse models as evidenced by significantly lower macroscopic and microscopic damage scores. AdipoRon exhibited anti-inflammatory effect by reduction in MPO activity and IL-1ß expression in the gastric tissue. Moreover, AdipoRon induced antioxidative action, as demonstrated with higher GSH levels, and increased SOD and GPX activity. CONCLUSIONS: Activation of AdipoR1 and AdipoR2 using AdipoRon reduced gastric lesions and enhanced cell response to oxidative stress. Our data suggest that AdipoR1 and AdipoR2 activation may be an attractive therapeutic strategy to inhibit development of gastric ulcers.


Asunto(s)
Omeprazol/administración & dosificación , Estrés Oxidativo/efectos de los fármacos , Piperidinas/administración & dosificación , Receptores de Adiponectina/agonistas , Úlcera Gástrica/tratamiento farmacológico , Administración Oral , Animales , Catalasa/metabolismo , Diclofenaco/efectos adversos , Modelos Animales de Enfermedad , Etanol/efectos adversos , Masculino , Ratones , Omeprazol/farmacología , Peroxidasa/metabolismo , Piperidinas/farmacología , Úlcera Gástrica/inducido químicamente , Úlcera Gástrica/metabolismo , Superóxido Dismutasa/metabolismo , Resultado del Tratamiento
3.
Postepy Biochem ; 65(4): 313-317, 2020 Jan 04.
Artículo en Polaco | MEDLINE | ID: mdl-31945286

RESUMEN

Inflammatory bowel diseases (IBD) are chronic conditions that lead to serious complications and act to the detriment of quality of patients' lives. Etiology of IBD has not been precisely determined but it is assumed that IBD is caused by genetic, immune and environmental factors. The main target in current IBD treatment is the induction and maintenance of remission. The most common strategy in IBD therapy is called "step-up" that is based on gradual introduction of stronger drugs. However, the latest research shows that "top down" strategy is more promising and can change the natural course of the disease. The aim of this article is to discuss both strategies and compare their effectiveness.


Asunto(s)
Enfermedad de Crohn/terapia , Enfermedad de Crohn/genética , Enfermedad de Crohn/inmunología , Humanos
4.
Curr Drug Targets ; 21(8): 792-806, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31854272

RESUMEN

Crohn's disease (CD) is an autoimmune disorder from the group of inflammatory bowel diseases. The etiology of CD is not clear; currently, the interaction between the genetic, immunological and environmental factors is assumed as the cause of the disease. Partial knowledge of those factors has led to the development of drugs, which control the clinical symptoms and improve the overall condition of the infected; the main objective of the modern therapeutic strategies is the induction and maintenance of remission. Among the wide range of available treatments, older generation molecules: mesalazine, corticosteroids and thiopurine derivatives as well as biological drugs and biosimilars can be distinguished. Moreover, some novel biologics and small molecule drugs have shown potential in CD clinical trials, providing safe and effective results. This article provides an overview of the achievements in the field of biologic therapy, its efficacy and safety with an indication of future directions in CD treatment.


Asunto(s)
Terapia Biológica/métodos , Enfermedad de Crohn/terapia , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Biosimilares Farmacéuticos/uso terapéutico , Enfermedad de Crohn/etiología , Humanos , Inhibidores de las Cinasas Janus/farmacología , Inhibidores de las Cinasas Janus/uso terapéutico , Inhibidores del Factor de Necrosis Tumoral/farmacología , Inhibidores del Factor de Necrosis Tumoral/uso terapéutico
5.
Front Oncol ; 9: 1087, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31681615

RESUMEN

Process of inflammation and complex interactions between immune and cancer cells within tumor microenvironment are known to drive and shape the outcome of the neoplastic disease. Recent studies increasingly show that ion channels can be used as potential targets to modulate immune response and to treat inflammatory disorders and cancer. The action of both innate and adaptive immune cells is tightly regulated by ionic signals provided by a network of distinct ion channels. TRPV1 channel, known as a capsaicin receptor, was recently documented to be expressed on the cells of the immune system but also aberrantly expressed in the several tumor types. It is activated by heat, protons, proinflammatory cytokines, and associated with pain and inflammation. TRPV1 channel is not only involved in calcium signaling fundamental for many cellular processes but also takes part in cell-environment crosstalk influencing cell behavior. Furthermore, in several studies, activation of TRPV1 by capsaicin was associated with anti-cancer effects. Therefore, TRPV1 provides a potential link between the process of inflammation, cancer and immunity, and offers new treatment possibilities. Nevertheless, in many cases, results regarding TRPV1 are contradictory and need further refinement. In this review we present the summary of the data related to the role of TRPV1 channel in the process of inflammation, cancer and immunity, limitations of the studies, and directions for future research.

6.
Nutrients ; 10(12)2018 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-30513813

RESUMEN

Gamma-oryzanol (GO) is a popular supplement for performance horses, dogs, and humans. Previous studies indicated that GO supplementation decreases creatine kinase activity and lactate level after exercise and may affect oxidative stress in Thoroughbred horses. GO may change genes expression in equine satellite cells (ESC). The purpose of this study was to evaluate the effect of GO on miRNA, gene expression, oxidative stress, and cell damage and viability in differentiating ESC pretreated with hydrogen peroxide (H2O2). ESCs were obtained from a young horse's skeletal muscle. ESCs were pre-incubated with GO (24 h) and then exposed to H2O2 for one hour. For the microRNA and gene expression assessment, the microarray technique was used. Identified miRNAs and genes were validated using real time-quantitative polymerase chain reaction. Several tests related to cell viability, cell damage, and oxidative stress were performed. The microarray analysis revealed differences in 17 miRNAs and 202 genes between GO-treated and control ESC. The tests related to apoptosis, cell viability, and oxidative stress showed that GO affects these processes to varying degrees. Our results suggest that GO can change miRNA and gene expression and may impact the processes involved in tissue repairing after an injury.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Perfilación de la Expresión Génica/veterinaria , Caballos , Peróxido de Hidrógeno/farmacología , Fenilpropionatos/farmacología , Células Satélite del Músculo Esquelético/fisiología , Animales , Apoptosis/efectos de los fármacos , Diferenciación Celular/genética , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica/métodos , Masculino , MicroARNs/análisis , Estrés Oxidativo/efectos de los fármacos , ARN Mensajero/análisis , Células Satélite del Músculo Esquelético/efectos de los fármacos , Análisis de Matrices Tisulares/métodos , Análisis de Matrices Tisulares/veterinaria
7.
Acta Vet Scand ; 60(1): 60, 2018 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-30305130

RESUMEN

Cancer immunotherapy is recently considered the most promising treatment for human patients with advanced tumors and could be effectively combined with conventional therapies such as chemotherapy or radiotherapy. Patients with hematological malignancies and melanoma have benefited greatly from immunotherapies such as, adoptive cell transfer therapy, experiencing durable remissions and prolonged survival. In the face of increasing enthusiasm for immunotherapy, particularly for the administration of tumor-specific T lymphocytes, the question arises whether this method could be employed to improve treatment outcomes for canine patients. It is warranted to determine whether veterinary clinical trials could support comparative oncology research and thus facilitate the development of new cell-based therapies for humans. Herein, we discuss adoptive transfer of T lymphocytes and lymphokine-activated cells for application in veterinary oncology, in the context of human medicine achievements. Furthermore, we discuss potential benefits of using domestic dog as a model for immunotherapy and its advantages for translational medicine. We also focus on an emerging genome-editing technology as a useful tool to improve a T cells' phenotype.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedades de los Perros/tratamiento farmacológico , Neoplasias/veterinaria , Traslado Adoptivo , Animales , Perros , Humanos , Inmunoterapia Adoptiva , Neoplasias/tratamiento farmacológico
8.
Cancer Res ; 78(14): 3888-3898, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29769201

RESUMEN

Adoptive T-cell transfer therapy is an FDA- approved treatment for leukemia that relies on the ex vivo expansion and reinfusion of a patient's immune cells, which can be engineered with a chimeric antigen receptor (CAR) for more efficient tumor recognition. Type 17 T cells, controlled transcriptionally by RORγ, have been reported to mediate potent antitumor effects superior to those observed with conventionally expanded T cells. Here, we demonstrate that addition of a synthetic, small-molecule RORγ agonist during ex vivo expansion potentiates the antitumor activity of human Th17 and Tc17 cells redirected with a CAR. Likewise, ex vivo use of this agonist bolstered the antitumor properties of murine tumor-specific CD4+ and CD8+ T cells. Expansion in the presence of the RORγ agonist enhanced IL17A production without compromising IFNγ secretion in vitroIn vivo, cytokine neutralization studies revealed that IFNγ and IL17A were required to regress murine melanoma tumors. The enhanced antitumor effect of RORγ agonist treatment was associated with recovery of more donor T cells in the tumor and spleen; these cells produced elevated levels of cytokines months after infusion and expressed markers of long-lived stem and central memory cells such as Tcf7 and CD62L. Conversely, untreated cells mainly exhibited effector phenotypes in the tumor. Cured mice previously treated with agonist-primed T cells were protected from tumor rechallenge. Collectively, our work reveals that in vitro treatment with a RORγ agonist generates potent antitumor Type 17 effector cells that persist as long-lived memory cells in vivoSignificance: RORγ agonists can be used in vitro during T-cell expansion to enhance the efficacy of adoptive cell therapy (e.g., CAR-T) and to provide long-term protection against tumors.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/14/3888/F1.large.jpg Cancer Res; 78(14); 3888-98. ©2018 AACR.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Células Th17/inmunología , Traslado Adoptivo/métodos , Animales , Línea Celular Tumoral , Citocinas/inmunología , Humanos , Inmunoterapia Adoptiva/métodos , Interferón gamma/inmunología , Interleucina-17 , Activación de Linfocitos/inmunología , Melanoma/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID
9.
Genes Nutr ; 13: 10, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29662554

RESUMEN

BACKGROUND: Skeletal muscle injury activates satellite cells to initiate processes of proliferation, differentiation, and hypertrophy in order to regenerate muscle fibers. The number of microRNAs and their target genes are engaged in satellite cell activation. ß-Hydroxy-ß-methylbutyrate (HMB) is known to prevent exercise-induced muscle damage. The purpose of this study was to evaluate the effect of HMB on miRNA and relevant target gene expression in differentiating equine satellite cells exposed to H2O2. We hypothesized that HMB may regulate satellite cell activity, proliferation, and differentiation, hence attenuate the pathological processes induced during an in vitro model of H2O2-related injury by changing the expression of miRNAs. METHODS: Equine satellite cells (ESC) were isolated from the samples of skeletal muscle collected from young horses. ESC were treated with HMB (24 h) and then exposed to H2O2 (1 h). For the microRNA and gene expression assessment microarrays, technique was used. Identified miRNAs and genes were validated using real-time qPCR. Cell viability, oxidative stress, and cell damage were measured using colorimetric method and flow cytometry. RESULTS: Analysis of miRNA and gene profile in differentiating ESC pre-incubated with HMB and then exposed to H2O2 revealed difference in the expression of 27 miRNAs and 4740 genes, of which 344 were potential target genes for identified miRNAs. Special attention was focused on differentially expressed miRNAs and their target genes involved in processes related to skeletal muscle injury. Western blot analysis showed protein protection in HMB-pre-treated group compared to control. The viability test confirmed that HMB enhanced cell survival after the hydrogen peroxide exposition. CONCLUSIONS: Our results suggest that ESC pre-incubated with HMB and exposed to H2O2 could affect expression on miRNA levels responsible for skeletal muscle development, cell proliferation and differentiation, and activation of tissue repair after injury. Enrichment analyses for targeted genes revealed that a large group of genes was associated with the regulation of signaling pathways crucial for muscle tissue development, protein metabolism, muscle injury, and regeneration, as well as with oxidative stress response.

10.
Nat Commun ; 8(1): 1961, 2017 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-29213079

RESUMEN

CD8+ T lymphocytes mediate potent immune responses against tumor, but the role of human CD4+ T cell subsets in cancer immunotherapy remains ill-defined. Herein, we exhibit that CD26 identifies three T helper subsets with distinct immunological properties in both healthy individuals and cancer patients. Although CD26neg T cells possess a regulatory phenotype, CD26int T cells are mainly naive and CD26high T cells appear terminally differentiated and exhausted. Paradoxically, CD26high T cells persist in and regress multiple solid tumors following adoptive cell transfer. Further analysis revealed that CD26high cells have a rich chemokine receptor profile (including CCR2 and CCR5), profound cytotoxicity (Granzyme B and CD107A), resistance to apoptosis (c-KIT and Bcl2), and enhanced stemness (ß-catenin and Lef1). These properties license CD26high T cells with a natural capacity to traffic to, regress and survive in solid tumors. Collectively, these findings identify CD4+ T cell subsets with properties critical for improving cancer immunotherapy.


Asunto(s)
Dipeptidil Peptidasa 4/inmunología , Dipeptidil Peptidasa 4/metabolismo , Neoplasias/inmunología , Subgrupos de Linfocitos T/inmunología , Traslado Adoptivo , Animales , Apoptosis , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Movimiento Celular/inmunología , Citocinas/inmunología , Dipeptidil Peptidasa 4/sangre , Modelos Animales de Enfermedad , Granzimas , Humanos , Inmunidad , Memoria Inmunológica , Inmunoterapia , Factor de Unión 1 al Potenciador Linfoide , Proteína 1 de la Membrana Asociada a los Lisosomas/inmunología , Melanoma/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias/patología , Proteínas Proto-Oncogénicas c-bcl-2 , Proteínas Proto-Oncogénicas c-kit , Linfocitos T Colaboradores-Inductores/inmunología , beta Catenina
11.
Front Immunol ; 8: 1221, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29033940

RESUMEN

Phosphatidylinositol-3-kinase p110δ (PI3Kδ) inhibition by Idelalisib (CAL-101) in hematological malignancies directly induces apoptosis in cancer cells and disrupts immunological tolerance by depleting regulatory T cells. Yet, little is known about the direct impact of PI3Kδ blockade on effector T cells from CAL-101 therapy. Herein, we demonstrate a direct effect of p110δ inactivation via CAL-101 on murine and human CD8+ T cells that promotes a strong undifferentiated phenotype (elevated CD62L/CCR7, CD127, and Tcf7). These CAL-101 T cells also persisted longer after transfer into tumor bearing mice in both the murine syngeneic and human xenograft mouse models. The less differentiated phenotype and improved engraftment of CAL-101 T cells resulted in stronger antitumor immunity compared to traditionally expanded CD8+ T cells in both tumor models. Thus, this report describes a novel direct enhancement of CD8+ T cells by a p110δ inhibitor that leads to markedly improved tumor regression. This finding has significant implications to improve outcomes from next generation cancer immunotherapies.

12.
J Immunol Res Ther ; 2(1): 68-79, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28825053

RESUMEN

Adoptive T cell transfer (ACT) can mediate objective responses in patients with advanced malignancies. There have been major advances in this field, including the optimization of the ex vivo generation of tumor-reactive lymphocytes to ample numbers for effective ACT therapy via the use of natural and artificial antigen presenting cells (APCs). Herein we review the basic properties of APCs and how they have been manufactured through the years to augment vaccine and T cell-based cancer therapies. We then discuss how these novel APCs impact the function and memory properties of T cells. Finally, we propose new ways to synthesize aAPCs to augment the therapeutic effectiveness of antitumor T cells for ACT therapy.

13.
DNA Cell Biol ; 36(7): 535-540, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28570836

RESUMEN

Hepatitis C virus (HCV) can infect extrahepatic tissues, including lymphocytes, creating reservoir of the virus. Moreover, HCV proteins can interact with DNA damage response proteins of infected cells. In this article we investigated the influence of the virus infection and a new ombitasvir/paritaprevir/ritonavir ± dasabuvir ± ribavirin (OBV/PTV/r ± DSV ± RBV) anti-HCV therapy on the PBMCs (peripheral blood mononuclear cells, mainly lymphocytes) DNA base excision repair (BER) system. BER protein activity was analyzed in the nuclear and mitochondrial extracts (NE and ME) of PBMC isolated from patients before and after therapy, and from subjects without HCV, using modeled double-strand DNA, with 2'-deoxyuridine substitution as the DNA damage. The NE and ME obtained from patients before therapy demonstrated lower efficacy of 2'-deoxyuridine removal and DNA repair polymerization than those of the control group or patients after therapy. Moreover, the extracts from the patients after therapy had similar activity to those from the control group. However, the efficacy of apurinic/apyrimidinic site excision in NE did not differ between the studied groups. We postulate that infection of lymphocytes by the HCV can lead to a decrease in the activity of BER enzymes. However, the use of novel therapy results in the improvement of glycosylase activity as well as the regeneration of endonuclease and other crucial repair enzymes.


Asunto(s)
Antivirales/farmacología , Núcleo Celular/efectos de los fármacos , Reparación del ADN , ADN/genética , Leucocitos Mononucleares/efectos de los fármacos , Mitocondrias/efectos de los fármacos , 2-Naftilamina , Anilidas/farmacología , Carbamatos/farmacología , Núcleo Celular/metabolismo , Núcleo Celular/virología , Ciclopropanos , ADN/metabolismo , Roturas del ADN de Doble Cadena , ADN Glicosilasas/genética , ADN Glicosilasas/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Desoxiuridina/metabolismo , Quimioterapia Combinada , Endonucleasas/genética , Endonucleasas/metabolismo , Expresión Génica , Hepacivirus/efectos de los fármacos , Hepacivirus/crecimiento & desarrollo , Hepatitis C Crónica/tratamiento farmacológico , Hepatitis C Crónica/virología , Interacciones Huésped-Patógeno , Humanos , Lactamas Macrocíclicas , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/virología , Compuestos Macrocíclicos/farmacología , Mitocondrias/metabolismo , Mitocondrias/virología , Imitación Molecular , Cultivo Primario de Células , Prolina/análogos & derivados , Ribavirina/farmacología , Ritonavir/farmacología , Sulfonamidas/farmacología , Uracilo/análogos & derivados , Uracilo/farmacología , Valina
14.
JCI Insight ; 2(8)2017 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-28422756

RESUMEN

ICOS costimulation generates Th17 cells with durable memory responses to tumor. Herein, we found that ICOS induces PI3K/p110δ/Akt and Wnt/ß-catenin pathways in Th17 cells. Coinhibiting PI3Kδ and ß-catenin altered the biological fate of Th17 cells. Th17 cells inhibited of both pathways expressed less RORγt, which, in turn, reduced their ability to secrete IL-17. Unexpectedly, these cells were more effective (than uninhibited cells) at regressing tumor when infused into mice, leading to long-term curative responses. PI3Kδ inhibition expanded precursor Th17 cells with a central memory phenotype that expressed nominal regulatory properties (low FoxP3), while ß-catenin inhibition enhanced Th17 multifunctionality in vivo. Remarkably, upon TCR restimulation, RORγt and IL-17 rebounded in Th17 cells treated with PI3Kδ and ß-catenin inhibitors. Moreover, these cells regained ß-catenin, Tcf7, and Akt expression, licensing them to secrete heightened IL-2, persist, and eradicate solid tumors without help from endogenous NK and CD8 T cells. This finding shines a light on ways to repurpose FDA-approved drugs to augment T cell-based cancer immunotherapies.

15.
JCI Insight ; 2(5): e90772, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28289713

RESUMEN

Adoptive immunotherapy for solid tumors relies on infusing large numbers of T cells to mediate successful antitumor responses in patients. While long-term rapid-expansion protocols (REPs) produce sufficient numbers of CD8+ T cells for treatment, they also cause decline in the cell's therapeutic fitness. In contrast, we discovered that IL-17-producing CD4+ T cells (Th17 cells) do not require REPs to expand 5,000-fold over 3 weeks. Also, unlike Th1 cells, Th17 cells do not exhibit hallmarks of senescence or apoptosis, retaining robust antitumor efficacy in vivo. Three-week-expanded Th17 cells eliminated melanoma as effectively as Th17 cells expanded for 1 week when infused in equal numbers into mice. However, treating mice with large recalcitrant tumors required the infusion of all cells generated after 2 or 3 weeks of expansion, while the cell yield obtained after 1-week expansion was insufficient. Long-term-expanded Th17 cells also protected mice from tumor rechallenge including lung metastasis. Importantly, 2-week-expanded human chimeric antigen receptor-positive (CAR+) Th17 cells also retained their ability to regress human mesothelioma, while CAR+ Th1 cells did not. Our results indicate that tumor-reactive Th17 cells are an effective cell therapy for cancer, remaining uncompromised when expanded for a long duration owing to their resistance to senescence.


Asunto(s)
Senescencia Celular , Inmunoterapia Adoptiva , Neoplasias Pulmonares/inmunología , Melanoma Experimental/inmunología , Mesotelioma/inmunología , Células Th17/citología , Células Th17/inmunología , Animales , Línea Celular Tumoral , Humanos , Memoria Inmunológica , Neoplasias Pulmonares/terapia , Melanoma Experimental/terapia , Mesotelioma/terapia , Mesotelioma Maligno , Ratones , Ratones Endogámicos C57BL
16.
Oncotarget ; 7(46): 75551-75560, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27705909

RESUMEN

Cancer including melanoma may be ''addicted" to double strand break (DSB) repair and targeting this process could sensitize them to the lethal effect of DNA damage. PARP1 exerts an important impact on DSB repair as it binds to both single- and double- strand breaks. PARP1 inhibitors might be highly effective drugs triggering synthetic lethality in patients whose tumors have germline or somatic defects in DNA repair genes. We hypothesized that PARP1-dependent synthetic lethality could be induced in melanoma cells displaying downregulation of DSB repair genes. We observed that PARP1 inhibitor olaparib sensitized melanomas with reduced expression of DNA ligase 4 (LIG4) to an alkylatimg agent dacarbazine (DTIC) treatment in vitro, while normal melanocytes remained intact. PARP1 inhibition caused accumulation of DSBs, which was associated with apoptosis in LIG4 deficient melanoma cells. Our hypothesis that olaparib is synthetic lethal with LIG4 deficiency in melanoma cells was supported by selective anti-tumor effects of olaparib used either alone or in combination with dacarbazine (DTIC) in LIG4 deficient, but not LIG4 proficient cells. In addition, olaparib combined with DTIC inhibited the growth of LIG4 deficient human melanoma xenografts. This work for the first time demonstrates the effectiveness of a combination of PARP1 inhibitor olaparib and alkylating agent DTIC for treating LIG4 deficient melanomas. In addition, analysis of the TCGA and transcriptome microarray databases revealed numerous individual melanoma samples potentially displaying specific defects in DSB repair pathways, which may predispose them to synthetic lethality triggered by PARP1 inhibitor combined with a cytotoxic drug.


Asunto(s)
Antineoplásicos/farmacología , ADN Ligasa (ATP)/genética , Melanoma/genética , Melanoma/metabolismo , Ftalazinas/farmacología , Piperazinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Mutaciones Letales Sintéticas , Animales , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , ADN Ligasa (ATP)/deficiencia , Reparación del ADN , Dacarbazina/farmacología , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Histonas/metabolismo , Humanos , Melanocitos/efectos de los fármacos , Melanocitos/metabolismo , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Mediators Inflamm ; 2016: 5230219, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27212807

RESUMEN

Recent studies indicate the critical role of tumour associated macrophages, tumour associated neutrophils, dendritic cells, T lymphocytes, and natural killer cells in tumourigenesis. These cells can have a significant impact on the tumour microenvironment via their production of cytokines and chemokines. Additionally, products secreted from all these cells have defined specific roles in regulating tumour cell proliferation, angiogenesis, and metastasis. They act in a protumour capacity in vivo as evidenced by the recent studies indicating that macrophages, T cells, and neutrophils may be manipulated to exhibit cytotoxic activity against tumours. Therefore therapy targeting these cells may be promising, or they may constitute drug or anticancer particles delivery systems to the tumours. Herein, we discussed all these possibilities that may be used in cancer treatment.


Asunto(s)
Neoplasias/terapia , Animales , Humanos , Macrófagos/metabolismo , Macrófagos/fisiología , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Neutrófilos/metabolismo , Neutrófilos/fisiología , Linfocitos T/metabolismo , Linfocitos T/fisiología , Microambiente Tumoral/inmunología
18.
J Inorg Biochem ; 159: 133-41, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26986980

RESUMEN

A series of arene ruthenium(II) complexes with the general formula [(η(6)-arene)Ru(L)X2] (where arene=p-cymene, benzene, hexamethylbenzene or mesitylene, L=aminoflavone or aminochromone derivatives and X=Cl, I) were synthesized and characterized by elemental analysis, MS, IR and (1)H NMR spectroscopy. The stability of the selected complexes was assessed by UV-Vis spectroscopy in 24-hour period. The lipophilicity of the synthesized complexes was determined by the shake-flask method, and their cytotoxicity evaluated in vitro on patient-derived melanoma populations. The most active complexes against melanoma cells contain 7-aminoflavone and 6-aminoflavone as a ligand. The relationship between the cytotoxicity of all the obtained compounds and their logP values was determined and briefly analyzed with two different patterns observed.


Asunto(s)
Antineoplásicos , Cromonas , Melanoma/tratamiento farmacológico , Octanoles/química , Rutenio , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Cromonas/síntesis química , Cromonas/química , Cromonas/farmacología , Citotoxinas/síntesis química , Citotoxinas/química , Citotoxinas/farmacología , Humanos , Melanoma/metabolismo , Rutenio/química , Rutenio/farmacología
19.
Cancer Immunol Immunother ; 65(3): 247-59, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26825102

RESUMEN

Cancer immunotherapy is one the most effective approaches for treating patients with tumors, as it bolsters the generation and persistence of memory T cells. In preclinical work, it has been reported that adoptively transferred CD4+ and CD8+ lymphocytes that secrete IL-17A (i.e., Th17 and Tc17 cells) regress tumors to a greater extent than IFN-γ(+)Th1 or Tc1 cells in vivo. Herein, we review the mechanisms underlying how infused Th17 and Tc17 cells regress established malignancies in clinically relevant mouse models of cancer. We also discuss how unique signaling cues--such as co-stimulatory molecules (ICOS and 41BB), cytokines (IL-12 and IL-23) or pharmaceutical reagents (Akt inhibitors, etc.)--can be exploited to bolster the therapeutic potential of IL-17(+) lymphocytes with an emphasis on using this knowledge to improve next-generation clinical trials for patients with cancer.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Interleucina-17/biosíntesis , Neoplasias/terapia , Humanos , Memoria Inmunológica , Inmunoterapia , Proteína Coestimuladora de Linfocitos T Inducibles/fisiología , Interleucina-12/fisiología , Neoplasias/inmunología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Células Th17/fisiología
20.
Oncoimmunology ; 5(12): e1254854, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28123897

RESUMEN

RORγt is the key transcription factor controlling the development and function of CD4+ Th17 and CD8+ Tc17 cells. Across a range of human tumors, about 15% of the CD4+ T cell fraction in tumor-infiltrating lymphocytes are RORγ+ cells. To evaluate the role of RORγ in antitumor immunity, we have identified synthetic, small molecule agonists that selectively activate RORγ to a greater extent than the endogenous agonist desmosterol. These RORγ agonists enhance effector function of Type 17 cells by increasing the production of cytokines/chemokines such as IL-17A and GM-CSF, augmenting expression of co-stimulatory receptors like CD137, CD226, and improving survival and cytotoxic activity. RORγ agonists also attenuate immunosuppressive mechanisms by curtailing Treg formation, diminishing CD39 and CD73 expression, and decreasing levels of co-inhibitory receptors including PD-1 and TIGIT on tumor-reactive lymphocytes. The effects of RORγ agonists were not observed in RORγ-/- T cells, underscoring the selective on-target activity of the compounds. In vitro treatment of tumor-specific T cells with RORγ agonists, followed by adoptive transfer to tumor-bearing mice is highly effective at controlling tumor growth while improving T cell survival and maintaining enhanced IL-17A and reduced PD-1 in vivo. The in vitro effects of RORγ agonists translate into single agent, immune system-dependent, antitumor efficacy when compounds are administered orally in syngeneic tumor models. RORγ agonists integrate multiple antitumor mechanisms into a single therapeutic that both increases immune activation and decreases immune suppression resulting in robust inhibition of tumor growth. Thus, RORγ agonists represent a novel immunotherapy approach for cancer.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...