Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Expert Rev Proteomics ; 15(3): 259-275, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29343144

RESUMEN

INTRODUCTION: Cancer is the second most common cause of death worldwide and its heterogeneity complicates therapy. Standard cytotoxic regiments disrupt rapidly dividing cells, regardless of their neoplastic status. The introduction of less toxic targeted therapies has partially contributed to the observed decrease in cancer-related mortality. Cell-surface proteins represent attractive targets for therapy, due to their easily-accessible localization and their involvement in essential signaling pathways, often dysregulated in cancer. Despite their clinical appeal, cell-surface proteins are often underrepresented in standard proteomic data sets, due to their poor solubility and lower expression levels compared to intracellular proteins. Areas covered: This review will summarize some of the available techniques for enriching the cell-surface proteome, and discuss their advantages, limitations and applicability to clinical sample-testing. Moreover, we discuss currently available strategies for the development of novel targeted therapies in cancer. Expert commentary: The interest in elucidating the cancer-associated surfaceome is growing and will likely benefit from recent advancements in instrument sensitivity, method development, and a growing body of high-quality proteomics databases. Multiomics studies, in combination with functional validations (e.g. dropout screens), and evaluation of the healthy surfaceome, will likely aid in the selection of relevant targets for future therapy development.


Asunto(s)
Antígenos de Neoplasias/química , Biomarcadores de Tumor/química , Terapia Molecular Dirigida/métodos , Proteómica/métodos , Animales , Antígenos de Neoplasias/metabolismo , Biomarcadores de Tumor/metabolismo , Humanos , Espectrometría de Masas/métodos
2.
Blood ; 127(21): 2575-86, 2016 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-26941401

RESUMEN

Herein we demonstrate that oncolytic herpes simplex virus-1 (HSV-1) potently activates human peripheral blood mononuclear cells (PBMCs) to lyse leukemic cell lines and primary acute myeloid leukemia samples, but not healthy allogeneic lymphocytes. Intriguingly, we found that UV light-inactivated HSV-1 (UV-HSV-1) is equally effective in promoting PBMC cytolysis of leukemic cells and is 1000- to 10 000-fold more potent at stimulating innate antileukemic responses than UV-inactivated cytomegalovirus, vesicular stomatitis virus, reovirus, or adenovirus. Mechanistically, UV-HSV-1 stimulates PBMC cytolysis of leukemic cells, partly via Toll-like receptor-2/protein kinase C/nuclear factor-κB signaling, and potently stimulates expression of CD69, degranulation, migration, and cytokine production in natural killer (NK) cells, suggesting that surface components of UV-HSV-1 directly activate NK cells. Importantly, UV-HSV-1 synergizes with interleukin-15 (IL-15) and IL-2 in inducing activation and cytolytic activity of NK cells. Additionally, UV-HSV-1 stimulates glycolysis and fatty acid oxidation-dependent oxygen consumption in NK cells, but only glycolysis is required for their enhanced antileukemic activity. Last, we demonstrate that T cell-depleted human PBMCs exposed to UV-HSV-1 provide a survival benefit in a murine xenograft model of human acute myeloid leukemia (AML). Taken together, our results support the preclinical development of UV-HSV-1 as an adjuvant, alone or in combination with IL-15, for allogeneic donor mononuclear cell infusions to treat AML.


Asunto(s)
Herpesvirus Humano 1/inmunología , Inmunidad Celular , Células Asesinas Naturales/inmunología , Leucemia/inmunología , Rayos Ultravioleta , Inactivación de Virus/efectos de la radiación , Degranulación de la Célula/inmunología , Movimiento Celular/inmunología , Femenino , Humanos , Interleucina-15/inmunología , Interleucina-2/inmunología , Células Jurkat , Masculino , FN-kappa B/inmunología , Proteína Quinasa C/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 2/inmunología
3.
Blood ; 126(16): 1874-5, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26472736

RESUMEN

In this issue of Blood, Ricciardi et al report a novel fatty acid oxidation (FAO) inhibitor, ST1326, that effectively inhibits proliferation, survival, and chemoresistance in leukemia cell lines and primary samples.


Asunto(s)
Carnitina O-Palmitoiltransferasa/antagonistas & inhibidores , Carnitina/análogos & derivados , Sistemas de Liberación de Medicamentos , Ácidos Grasos/metabolismo , Leucemia Mieloide Aguda/tratamiento farmacológico , Proteínas de Neoplasias/antagonistas & inhibidores , Humanos
4.
Haematologica ; 102(9): 1537-1548, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28659338

RESUMEN

The bone marrow microenvironment is known to provide a survival advantage to residual acute myeloid leukemia cells, possibly contributing to disease recurrence. The mechanisms by which stroma in the microenvironment regulates leukemia survival remain largely unknown. Using reverse-phase protein array technology, we profiled 53 key protein molecules in 11 signaling pathways in 20 primary acute myeloid leukemia samples and two cell lines, aiming to understand stroma-mediated signaling modulation in response to the targeted agents temsirolimus (MTOR), ABT737 (BCL2/BCL-XL), and Nutlin-3a (MDM2), and to identify the effective combination therapy targeting acute myeloid leukemia in the context of the leukemia microenvironment. Stroma reprogrammed signaling networks and modified the sensitivity of acute myeloid leukemia samples to all three targeted inhibitors. Stroma activated AKT at Ser473 in the majority of samples treated with single-agent ABT737 or Nutlin-3a. This survival mechanism was partially abrogated by concomitant treatment with temsirolimus plus ABT737 or Nutlin-3a. Mapping the signaling networks revealed that combinations of two inhibitors increased the number of affected proteins in the targeted pathways and in multiple parallel signaling, translating into facilitated cell death. These results demonstrated that a mechanism-based selection of combined inhibitors can be used to guide clinical drug selection and tailor treatment regimens to eliminate microenvironment-mediated resistance in acute myeloid leukemia.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Leucemia Mieloide Aguda/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Animales , Línea Celular , Femenino , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Masculino , Ratones , Células Tumorales Cultivadas
5.
Blood ; 122(20): 3398-400, 2013 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-24235130

RESUMEN

In this issue of Blood, Willems et al describe the dependence of acute myeloid leukemia (AML) cells on glutamine for maintaining protein synthesis downstream of mammalian target of rapamycin (mTOR) and show that the enzyme asparaginase can be used to target this dependence. Using various AML cell lines, primary samples, and CD341 stem cells from healthy donors, the authors support the notion that asparaginase may offer a therapeutic benefit in AML­not from its well-known enzymatic activity, but from its "off-target" effects on glutamine levels that result in inhibition of downstream mTOR signaling, inhibition of protein synthesis, and ultimately loss of viability.


Asunto(s)
Glutamina/antagonistas & inhibidores , Leucemia Mieloide Aguda/tratamiento farmacológico , Animales , Femenino , Humanos , Masculino
6.
BMC Musculoskelet Disord ; 16: 153, 2015 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-26092588

RESUMEN

BACKGROUND: Platelet-rich plasma (PRP) preparations are a common treatment in osteoarthritis (OA) and inflammatory synovitis. However, there is ambiguity regarding the ideal concentration of leukocytes and platelets in these preparations necessary to induce an adequate anti-inflammatory and anabolic response in joint tissues, such as the synovial membrane. This research aimed to study, in normal synovial membrane explants (SME) challenged with lipopolysaccharide (LPS), the temporal effects (at 48 and 96h) of leukocyte- and platelet-rich gel (L-PRG) and pure platelet-rich gel (P-PRG) supernatants on the production and degradation of platelet associated growth factors (GF) (platelet derived GF isoform BB (PDGF-BB) and transforming growth factor beta-1 (TGF-ß1)), pro-inflammatory (tumour necrosis factor alpha (TNF-α)) and anti-inflammatory cytokines (interleukin 4 (IL-4) and IL-1 receptor antagonists (IL-1ra)) and hyaluronan (HA). METHODS: Synovial membrane explants (SMEs) from 6 horses were challenged with LPS and cultured for 96h with L-PRG and P-PRG supernatants at concentrations of 25 and 50 %, respectively. The SME culture medium was changed every 48h and used for determination by ELISA of PDGF-BB, TGF-ß1, TNF-α, IL-4, IL-1ra and HA. These molecules were also determined in synovial fluid from the horses. RESULTS: Both the 25 and 50 % PRG supernatants produced a molecular profile in the culture media unlike that of the SME challenged with LPS only. They presented GF, cytokine and HA concentrations very near to the concentrations of these molecules in normal synovial fluid when compared with the SME control groups (either with LPS or without LPS). However, in comparison with the rest of the SME treated groups, the 25 % L-PRG produced the most IL-1ra, and the 50 % P-PRG induced the sustained production of IL-4 and HA. CONCLUSIONS: These in vitro findings suggest that anabolic and anti-inflammatory joint responses depend on the leukocyte and platelet concentration of the PRP preparation and on the volume of this substance injected. Moreover, it is possible, that leukoreduced PRP preparations are more effective for the medical treatment of patients with OA and inflammatory synovitis.


Asunto(s)
Plaquetas/metabolismo , Citocinas/metabolismo , Ácido Hialurónico/metabolismo , Mediadores de Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Lipopolisacáridos/farmacología , Plasma Rico en Plaquetas/metabolismo , Membrana Sinovial/efectos de los fármacos , Animales , Plaquetas/inmunología , Fraccionamiento Celular , Medios de Cultivo/metabolismo , Geles , Caballos , Leucocitos/efectos de los fármacos , Leucocitos/inmunología , Leucocitos/metabolismo , Plasma Rico en Plaquetas/inmunología , Líquido Sinovial/inmunología , Líquido Sinovial/metabolismo , Membrana Sinovial/inmunología , Membrana Sinovial/metabolismo , Factores de Tiempo , Técnicas de Cultivo de Tejidos
7.
Carcinogenesis ; 35(10): 2291-9, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25023988

RESUMEN

We recently demonstrated that both murine and human carcinomas grow significantly slower in mice on low carbohydrate (CHO), high protein diets than on isocaloric Western diets and that a further reduction in tumor growth rates occur when the low CHO diets are combined with the cyclooxygenase-2 inhibitor, celecoxib. Following upon these studies, we asked herein what effect low CHO, high protein diets, with or without celecoxib, might have on tumor metastasis. In the highly metastatic 4T1 mouse mammary tumor model, a 15% CHO, high protein diet supplemented with celecoxib (1 g/kg chow) markedly reduced lung metastases. Moreover, in longer-term studies using male Transgenic Adenocarcinoma of the Mouse Prostate mice, which are predisposed to metastatic prostate cancer, the 15% CHO diet, with and without celecoxib (0.3 g/kg chow), gave the lowest incidence of metastases, but a more moderate 25% CHO diet containing celecoxib led to the best survival. Metabolic studies with 4T1 tumors suggested that the low CHO, high protein diets may be forcing tumors to become dependent on amino acid catabolism for survival/growth. Taken together, our results suggest that a combination of a low CHO, high protein diet with celecoxib substantially reduces metastasis.


Asunto(s)
Dieta Baja en Carbohidratos , Proteínas en la Dieta/farmacología , Metástasis de la Neoplasia/tratamiento farmacológico , Pirazoles/farmacología , Sulfonamidas/farmacología , Animales , Celecoxib , Dietoterapia/métodos , Modelos Animales de Enfermedad , Neoplasias Pulmonares/dietoterapia , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Metástasis de la Neoplasia/terapia , Neoplasias de la Próstata/dietoterapia , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología
8.
Cancer Cell ; 10(5): 375-88, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17097560

RESUMEN

BCL-2 proteins are critical for cell survival and are overexpressed in many tumors. ABT-737 is a small-molecule BH3 mimetic that exhibits single-agent activity against lymphoma and small-cell lung cancer in preclinical studies. We here report that ABT-737 effectively kills acute myeloid leukemia blast, progenitor, and stem cells without affecting normal hematopoietic cells. ABT-737 induced the disruption of the BCL-2/BAX complex and BAK-dependent but BIM-independent activation of the intrinsic apoptotic pathway. In cells with phosphorylated BCL-2 or increased MCL-1, ABT-737 was inactive. Inhibition of BCL-2 phosphorylation and reduction of MCL-1 expression restored sensitivity to ABT-737. These data suggest that ABT-737 could be a highly effective antileukemia agent when the mechanisms of resistance identified here are considered.


Asunto(s)
Apoptosis/fisiología , Compuestos de Bifenilo , Resistencia a Antineoplásicos/fisiología , Leucemia Mieloide Aguda , Nitrofenoles , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Sulfonamidas , Animales , Compuestos de Bifenilo/metabolismo , Compuestos de Bifenilo/uso terapéutico , Línea Celular , Dimerización , Células Madre Hematopoyéticas/fisiología , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Ratones , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Proteínas de Neoplasias/metabolismo , Nitrofenoles/metabolismo , Nitrofenoles/uso terapéutico , Piperazinas/metabolismo , Piperazinas/uso terapéutico , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-bcl-2/química , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Sulfonamidas/metabolismo , Sulfonamidas/uso terapéutico , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/química , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
9.
Blood Adv ; 8(8): 1869-1879, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38330193

RESUMEN

ABSTRACT: Fc gamma receptor (FcγR) IIIA is an important receptor for immunoglobulin G (IgG) and is involved in immune defense mechanisms as well as tissue destruction in some autoimmune diseases including immune thrombocytopenia (ITP). FcγRIIIA on macrophages can trigger phagocytosis of IgG-sensitized platelets, and prior pilot studies observed blockade of FcγRIIIA increased platelet counts in patients with ITP. Unfortunately, although blockade of FcγRIIIA in patients with ITP increased platelet counts, its engagement by the blocking antibody drove serious adverse inflammatory reactions. These adverse events were postulated to originate from the antibody's Fc and/or bivalent nature. The blockade of human FcγRIIIA in vivo with a monovalent construct lacking an active Fc region has not yet been achieved. To effectively block FcγRIIIA in vivo, we developed a high affinity monovalent single-chain variable fragment (scFv) that can bind and block human FcγRIIIA. This scFv (17C02) was expressed in 3 formats: a monovalent fusion protein with albumin, a 1-armed human IgG1 antibody, and a standard bivalent mouse (IgG2a) antibody. Both monovalent formats were effective in preventing phagocytosis of ITP serum-sensitized human platelets. In vivo studies using FcγR-humanized mice demonstrated that both monovalent therapeutics were also able to increase platelet counts. The monovalent albumin fusion protein did not have adverse event activity as assessed by changes in body temperature, whereas the 1-armed antibody induced some changes in body temperature even though the Fc region function was impaired by the Leu234Ala and Leu235Ala mutations. These data demonstrate that monovalent blockade of human FcγRIIIA in vivo can potentially be a therapeutic strategy for patients with ITP.


Asunto(s)
Púrpura Trombocitopénica Idiopática , Trombocitopenia , Humanos , Ratones , Animales , Púrpura Trombocitopénica Idiopática/tratamiento farmacológico , Receptores de IgG/metabolismo , Modelos Animales de Enfermedad , Inmunoglobulina G/uso terapéutico , Albúminas/uso terapéutico
10.
Carcinogenesis ; 34(12): 2870-9, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23803693

RESUMEN

Metformin is a widely used antidiabetic drug, and epidemiology studies for pancreatic and other cancers indicate that metformin exhibits both chemopreventive and chemotherapeutic activities. Several metformin-induced responses and genes are similar to those observed after knockdown of specificity protein (Sp) transcription factors Sp1, Sp3 and Sp4 by RNA interference, and we hypothesized that the mechanism of action of metformin in pancreatic cancer cells was due, in part, to downregulation of Sp transcription factors. Treatment of Panc1, L3.6pL and Panc28 pancreatic cancer cells with metformin downregulated Sp1, Sp3 and Sp4 proteins and several pro-oncogenic Sp-regulated genes including bcl-2, survivin, cyclin D1, vascular endothelial growth factor and its receptor, and fatty acid synthase. Metformin induced proteasome-dependent degradation of Sps in L3.6pL and Panc28 cells, whereas in Panc1 cells metformin decreased microRNA-27a and induced the Sp repressor, ZBTB10, and disruption of miR-27a:ZBTB10 by metformin was phosphatase dependent. Metformin also inhibited pancreatic tumor growth and downregulated Sp1, Sp3 and Sp4 in tumors in an orthotopic model where L3.6pL cells were injected directly into the pancreas. The results demonstrate for the first time that the anticancer activities of metformin are also due, in part, to downregulation of Sp transcription factors and Sp-regulated genes.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Metformina/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Factores de Transcripción Sp/genética , Antineoplásicos/farmacología , Línea Celular Tumoral , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Neoplasias Pancreáticas/metabolismo , Factores de Transcripción Sp/metabolismo
11.
Cytotherapy ; 15(1): 20-32, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23260083

RESUMEN

BACKGROUND AIMS: Many ovarian cancers originate from ovarian surface epithelium, where they develop from cysts intermixed with stroma. The stromal layer is critical to the progression and survival of the neoplasm and consequently is recruited into the tumor microenvironment. METHODS: Using both syngeneic mouse tumors (ID8-R) and human xenograft (OVCAR3, SKOV3) tumor models, we first confirmed that intraperitoneally injected circulating mesenchymal stem cells (MSCs) could target, preferentially engraft and differentiate into α-smooth muscle actin-positive myofibroblasts, suggesting their role as "reactive stroma" in ovarian carcinoma development and confirming their potential as a targeted delivery vehicle for the intratumoral production of interferon-ß (IFN-ß). Mice with ovarian carcinomas then received weekly intraperitoneal injections of IFN-ß expressing MSCs. RESULTS: Intraperitoneal injections of IFN-ß expressing MSCs resulted in complete eradication of tumors in 70% of treated OVCAR3 mice (P = 0.004) and an increased survival of treated SKOV3 mice compared with controls (P = 0.01). Similar tumor growth control was observed using murine IFN-ß delivered by murine MSCs in ID8-R ovarian carcinoma. As a potential mechanism of tumor killing, MSCs produced IFN-ß-induced caspase-dependent tumor cell apoptosis. CONCLUSIONS: Our results demonstrate that ovarian carcinoma engrafts MSCs to participate in myofibrovascular networks and that IFN-ß produced by MSCs intratumorally modulates tumor kinetics, resulting in prolonged survival.


Asunto(s)
Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Neoplasias Ováricas/terapia , Animales , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Femenino , Terapia Genética/métodos , Humanos , Inmunohistoquímica , Interferón beta/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Ensayos Antitumor por Modelo de Xenoinjerto
12.
BMC Vet Res ; 9: 29, 2013 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-23402541

RESUMEN

BACKGROUND: There is no information on the effects of the breed, gender and age on the cellular content and growth factor (GF) release from equine pure-platelet rich plasma (P-PRP) and pure-platelet rich gel (P-PRG). The objectives of this study were: 1) to compare the cellular composition of P-PRP with whole blood and platelet poor plasma (PPP); 2) to compare the concentration of transforming GF beta 1 (TGF-ß1) and platelet derived GF isoform BB (PDGF-BB) between P-PRP treated with non-ionic detergent (P-PRP+NID), P-PRG (activated with calcium gluconate -CG-), PPP+NID, PPP gel (PPG), and plasma and; 3) to evaluate and to correlate the effect of the breed, gender and age on the cellular and GF concentration for each blood component. Forty adult horses, 20 Argentinean Creole Horses (ACH) and, 20 Colombian Creole Horses (CCH) were included. Data were analyzed by parametric (i.e.: t-test, one way ANOVA) and non parametric (Kruskal-Wallis test, Wilcoxon test) tests. Correlation analysis was also performed by using the Spearman and Pearson tests. A p ≤ 0.05 was set as significant for all tests. All the blood components were compared for platelet (PLT), leukocyte (WBC), TGF-ß1 and PDGF-BB concentrations. The effect of the breed, gender and age on these variables was analyzed. A P ≤ 0.05 was accepted as significant for all the tests. RESULTS: PLT counts were 1.8 and 0.6 times higher in P-PRP than in whole blood and PPP, respectively; WBC counts were 0.5 and 0.1 times lower in P-PRP, in comparison with whole blood and PPP, respectively. TGF-ß1 and PDGF-BB concentrations were 2.3 and 262 times higher, respectively, in P-PRG than in plasma, and 0.59 and 0.48 times higher, respectively, in P-PRG than in PPG. P-PRG derived from CCH females or young horses presented significantly (P < 0.001) higher PDGF-BB concentrations than P-PRG derived from ACH males or older horses. CONCLUSIONS: Our results indicated that P-PRP obtained by a manual method was affected by intrinsic factors such as the breed, gender and age. Equine practitioners should be aware that cellular and GF release from P-PRP/P-PRG could change according with the intrinsic variables associated with a patient in particular.


Asunto(s)
Plaquetas/metabolismo , Caballos/sangre , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Plasma Rico en Plaquetas/citología , Factores de Edad , Animales , Plaquetas/fisiología , Femenino , Caballos/fisiología , Recuento de Leucocitos/veterinaria , Masculino , Recuento de Plaquetas/veterinaria , Factor de Crecimiento Derivado de Plaquetas/fisiología , Plasma Rico en Plaquetas/metabolismo , Plasma Rico en Plaquetas/fisiología , Isoformas de Proteínas , Factores Sexuales , Especificidad de la Especie , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/fisiología
13.
Mol Ther Oncolytics ; 28: 334-348, 2023 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-36938544

RESUMEN

VG2025 is a recombinant oncolytic herpes simplex virus type 1 (HSV-1) that uses transcriptional and translational dual regulation (TTDR) of critical viral genes to enhance virus safety and promote tumor-specific virus replication without reducing virulence. The TTDR platform is based on transcriptional control of the essential HSV-1 immediate-early protein ICP27 using a tumor-specific carcinoembryonic antigen (CEA) promoter, coupled with translational control of the neurovirulence factor ICP34.5 using multiple microRNA (miR)-binding sites. VG2025 further incorporates IL-12 and the IL-15/IL-15 receptor alpha subunit complex to enhance the antitumor and immune stimulatory properties of oncolytic HSVs. The TTDR strategy was verified in vitro and shown to be highly selective. Strong in vivo antitumor efficacy was observed following both intratumoral and intravenous administration. Clear abscopal and immune memory effects were also evident, indicating a robust antitumor immune response. Gene expression profiling of treated tumors revealed increased immune cell infiltration and activation of multiple immune-signaling pathways when compared with the backbone virus. Absence of neurotoxicity was verified in mice and in rhesus monkeys. Taken together, the enhanced tumor clearance, excellent safety profile, and positive correlation between CEA levels and viral replication efficiency may provide an opportunity for using biomarker-based precision medicine in oncolytic virotherapy.

14.
Nutr Neurosci ; 15(1): 1-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22305647

RESUMEN

Polyphenols are secondary metabolites with antioxidant properties and are abundant in the diet. Fruits, vegetables, herbs, and various drinks (tea, wine, and juices) are all sources of these molecules. Despite their abundance, investigations into the benefits of polyphenols in human health have only recently begun. Phenolic compounds have received increasing interest because of numerous epidemiological studies. These studies have suggested associations between the consumption of polyphenol-rich aliments and the prevention of chronic diseases, such as cancer, cardiovascular diseases, and neurodegenerative diseases. More specifically, in the last 10 years literature on the neuroprotective effects of a polyphenol-rich diet has grown considerably. It has been demonstrated, in various cell culture and animal models, that these metabolites are able to protect neuronal cells by attenuating oxidative stress and damage. However, it remains unclear as to how these compounds reach the brain, what concentrations are necessary, and what biologically active forms are needed to exert beneficial effects. Therefore, further research is needed to identify the molecular pathways and intracellular targets responsible for polyphenol's neuroprotective effects. The aim of this paper is to present various well-known dietary polyphenols and their mechanisms of neuroprotection with an emphasis on Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Antioxidantes/metabolismo , Dieta , Fármacos Neuroprotectores/metabolismo , Enfermedad de Parkinson/metabolismo , Enfermedad de Alzheimer/prevención & control , Esclerosis Amiotrófica Lateral/prevención & control , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Humanos , Estrés Oxidativo/efectos de los fármacos , Enfermedad de Parkinson/prevención & control , Polifenoles/metabolismo
15.
Nutr Neurosci ; 15(3): 120-6, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22732354

RESUMEN

Parkinson's disease (PD) is a neurodegenerative movement disorder characterized by the degeneration and progressive loss of dopaminergic neurons in the substantia nigra pars compacta. It has been suggested that oxidative stress plays a role in the etiology and progression of PD. For instance, low levels of endogenous antioxidants, increased reactive species, augmented dopamine oxidation, and high iron levels have been found in brains from PD patients. In vitro and in vivo studies of Parkinson models evaluating natural and endogenous antioxidants such as polyphenols, coenzyme Q10, and vitamins A, C, and E have shown protective effects against oxidative-induced neuronal death. In this paper, we will review the mechanisms by which polyphenols and endogenous antioxidants can produce protection. Some of the mechanisms reviewed include: scavenging nitrogen and oxygen reactive species, regulation of signaling pathways associated with cell survival and inflammation, and inhibition of synphilin-1 and alpha-synuclein aggregation.


Asunto(s)
Antioxidantes/farmacología , Enfermedad de Parkinson/tratamiento farmacológico , Polifenoles/farmacología , Animales , Ácido Ascórbico/farmacología , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Muerte Celular/efectos de los fármacos , Dopamina/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Humanos , Inflamación/metabolismo , Inflamación/prevención & control , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Enfermedad de Parkinson/fisiopatología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , Ubiquinona/análogos & derivados , Ubiquinona/farmacología , Vitamina A/farmacología , Vitamina E/farmacología , alfa-Sinucleína/antagonistas & inhibidores , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
16.
Viruses ; 14(11)2022 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-36366425

RESUMEN

Oncolytic viruses (OVs) can specifically replicate in the host and cause cancer cell lysis while inducing an antitumor immune response. The aim of this study is to investigate the impact of either pre-existing immunity against herpes simplex virus type-1 (HSV-1) or multicycle treatment with OVs on anticancer efficacy of VG161, an HSV-1 OV in phase 2 clinical trial. VG161 efficacy was tested in CT26 mouse models by comparing the efficacy and immune response in naïve mice or in mice that were immunized with VG161. Moreover, VG161 efficacy in HLA-matched CD34+ humanized intrahepatic cholangiocarcinoma (ICC) patient-derived xenograft (PDX) models was also tested in multicycle treatment and was compared to standard chemotherapy for this type of cancer (gemcitabine). The HSV-1-immunized mice significantly inhibited tumor growth in VG161-treated mice compared to control naïve treated mice. RNA expression profiling and ELISPOT analyses indicated changes in the tumor's immune profile in the immunized and treated group compared to naïve and treated mice, as well as enhanced T cell function depicted by higher numbers of tumor specific lymphocytes, which was enhanced by immunization. In the ICC PDX model, repeated treatment of VG161 with 2 or 3 cycles seemed to increase the anticancer efficacy of VG161. In conclusion, the anticancer efficacy of VG161 can be enhanced by pre-immunization with HSV-1 and multicycle administration when the virus is given intratumorally, indicating that pre-existing antiviral immunity might enhance OV-induced antitumor immunity. Our results suggest potential clinical benefits of HSV-1-based OV therapy in HSV-1-seropositive patients and multicycle administration of VG161 for long-term maintenance treatment.


Asunto(s)
Herpesvirus Humano 1 , Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Humanos , Ratones , Animales , Virus Oncolíticos/fisiología , Herpesvirus Humano 1/genética , Neoplasias/terapia , Inmunidad , Modelos Animales de Enfermedad , Viroterapia Oncolítica/métodos
17.
Front Oncol ; 12: 856424, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35600398

RESUMEN

Podocalyxin (Podxl) is a CD34-related cell surface sialomucin that is normally highly expressed by adult vascular endothelia and kidney podocytes where it plays a key role in blocking adhesion. Importantly, it is also frequently upregulated on a wide array of human tumors and its expression often correlates with poor prognosis. We previously showed that, in xenograft studies, Podxl plays a key role in metastatic disease by making tumor initiating cells more mobile and invasive. Recently, we developed a novel antibody, PODO447, which shows exquisite specificity for a tumor-restricted glycoform of Podxl but does not react with Podxl expressed by normal adult tissue. Here we utilized an array of glycosylation defective cell lines to further define the PODO447 reactive epitope and reveal it as an O-linked core 1 glycan presented in the context of the Podxl peptide backbone. Further, we show that when coupled to monomethyl auristatin E (MMAE) toxic payload, PODO447 functions as a highly specific and effective antibody drug conjugate (ADC) in killing ovarian, pancreatic, glioblastoma and leukemia cell lines in vitro. Finally, we demonstrate PODO447-ADCs are highly effective in targeting human pancreatic and ovarian tumors in xenografted NSG and Nude mouse models. These data reveal PODO447-ADCs as exquisitely tumor-specific and highly efficacious immunotherapeutic reagents for the targeting of human tumors. Thus, PODO447 exhibits the appropriate characteristics for further development as a targeted clinical immunotherapy.

18.
Blood ; 113(7): 1504-12, 2009 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-18957686

RESUMEN

The CXCR4/SDF-1 axis has been studied extensively because of its role in development and hematopoiesis. In acute myeloid leukemia (AML), elevated expression of CXCR4 has been shown to correlate with shortened survival. Hy-poxia increases CXCR4 in several tumor models, but the impact of reduced O(2) partial pressure (pO(2)) on expression and biologic function of CXCR4 in AML is unknown. We determined pO(2) in bone marrows of AML patients as 6.1% (+/-1.7%). At this pO(2), CXCR4 surface and total expression were up-regulated within 10 hours in leukemic cell lines and patient samples as shown by Western blotting, fluorescence-activated cell sorting, and microscopy. Interestingly, hypoxic cells failed to internalize CXCR4 in response to SDF-1, and upon reoxygenation at 21% O(2), surface and total expression of CXCR4 rapidly decreased independent of adenosine triphosphate or proteasome activity. Instead, increased pO(2) led to alteration of lipid rafts by cholesterol depletion and structural changes and was associated with increased shedding of CXCR4-positive microparticles, suggesting a novel mechanism of CXCR4 regulation. Given the importance of CXCR4 in cell signaling, survival, and adhesion in leukemia, the results suggest that pO(2) be considered a critical variable in conducting and interpreting studies of CXCR4 expression and regulation in leukemias.


Asunto(s)
Hipoxia/metabolismo , Leucemia Mieloide Aguda/metabolismo , Oxígeno/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Médula Ósea/metabolismo , Médula Ósea/patología , Membrana Celular/fisiología , Quimiocina CXCL12/metabolismo , Regulación Leucémica de la Expresión Génica , Humanos , Hipoxia/complicaciones , Leucemia Mieloide Aguda/complicaciones , Leucemia Mieloide Aguda/patología , Presión Parcial , Transducción de Señal/fisiología , Células U937
19.
Blood ; 113(24): 6215-24, 2009 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-18955566

RESUMEN

SDF-1alpha/CXCR4 signaling plays a key role in leukemia/bone marrow microenvironment interactions. We previously reported that bone marrow-derived stromal cells inhibit chemotherapy-induced apoptosis in acute myeloid leukemia (AML). Here we demonstrate that the CXCR4 inhibitor AMD3465 antagonized stromal-derived factor 1alpha (SDF-1alpha)-induced and stroma-induced chemotaxis and inhibited SDF-1alpha-induced activation of prosurvival signaling pathways in leukemic cells. Further, CXCR4 inhibition partially abrogated the protective effects of stromal cells on chemotherapy-induced apoptosis in AML cells. Fetal liver tyrosine kinase-3 (FLT3) gene mutations activate CXCR4 signaling, and coculture with stromal cells significantly diminished antileukemia effects of FLT3 inhibitors in cells with mutated FLT3. Notably, CXCR4 inhibition increased the sensitivity of FLT3-mutated leukemic cells to the apoptogenic effects of the FLT3 inhibitor sorafenib. In vivo studies demonstrated that AMD3465, alone or in combination with granulocyte colony-stimulating factor, induced mobilization of AML cells and progenitor cells into circulation and enhanced antileukemic effects of chemotherapy and sorafenib, resulting in markedly reduced leukemia burden and prolonged survival of the animals. These findings indicate that SDF-1alpha/CXCR4 interactions contribute to the resistance of leukemic cells to signal transduction inhibitor- and chemotherapy-induced apoptosis in systems mimicking the physiologic microenvironment. Disruption of these interactions with CXCR4 inhibitors represents a novel strategy of sensitizing leukemic cells by targeting their protective bone marrow microenvironment.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Leucemia Experimental/tratamiento farmacológico , Leucemia Mieloide Aguda/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Receptores CXCR4/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Quimiotaxis/efectos de los fármacos , Citometría de Flujo , Humanos , Técnicas para Inmunoenzimas , Leucemia Experimental/metabolismo , Leucemia Experimental/patología , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos BALB C , Mutación/genética , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/metabolismo
20.
Mol Pharmacol ; 78(2): 226-36, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20488920

RESUMEN

The anticancer agent 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid (CDDO) and its methyl ester (CDDO-Me) typically induce a broad spectrum of growth-inhibitory, proapoptotic, and antiangiogenic responses. Treatment of Panc1, Panc28, and L3.6pL pancreatic cancer cells with low micromolar concentrations of CDDO or CDDO-Me resulted in growth inhibition, induction of apoptosis, and down-regulation of cyclin D1, survivin, vascular endothelial growth factor (VEGF), and its receptor (VEGFR2). RNA interference studies indicate that these repressed genes are regulated by specificity protein (Sp) transcription factors Sp1, Sp3, and Sp4, and Western blot analysis of lysates from pancreatic cancer cells treated with CDDO and CDDO-Me shows for the first time that both compounds decreased the expression of Sp1, Sp3, and Sp4. Moreover, CDDO-Me (7.5 mg/kg/day) also inhibited pancreatic human L3.6pL tumor growth and down-regulated Sp1, Sp3, and Sp4 in tumors using an orthotopic pancreatic cancer model. CDDO-Me also induced reactive oxygen species (ROS) and decreased mitochondrial membrane potential (MMP) in Panc1 and L3.6pL cells, and cotreatment with antioxidants (glutathione and dithiothreitol) blocked the formation of ROS, reversed the loss of MMP, and inhibited down-regulation of Sp1, Sp3, and Sp4. Repression of Sp and Sp-dependent genes by CDDO-Me was due to the down-regulation of microRNA-27a and induction of zinc finger and BTB domain containing 10 (ZBTB10), an Sp repressor, and these responses were also reversed by antioxidants. Thus, the anticancer activity of CDDO-Me is due, in part, to activation of ROS, which in turn targets the microRNA-27a:ZBTB10-Sp transcription factor axis. This results in decreased expression of Sp-regulated genes, growth inhibition, induction of apoptosis, and antiangiogenic responses.


Asunto(s)
MicroARNs/metabolismo , Ácido Oleanólico/análogos & derivados , Neoplasias Pancreáticas/patología , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Western Blotting , Línea Celular Tumoral , Proliferación Celular , Cartilla de ADN , Humanos , Masculino , Ratones , Ratones Desnudos , Ácido Oleanólico/farmacología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Reacción en Cadena de la Polimerasa , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA