Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Nat Commun ; 14(1): 5727, 2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37714830

RESUMEN

The poor efficacy of chimeric antigen receptor T-cell therapy (CAR T) for solid tumors is due to insufficient CAR T cell tumor infiltration, in vivo expansion, persistence, and effector function, as well as exhaustion, intrinsic target antigen heterogeneity or antigen loss of target cancer cells, and immunosuppressive tumor microenvironment (TME). Here we describe a broadly applicable nongenetic approach that simultaneously addresses the multiple challenges of CAR T as a therapy for solid tumors. The approach reprograms CAR T cells by exposing them to stressed target cancer cells which have been exposed to the cell stress inducer disulfiram (DSF) and copper (Cu)(DSF/Cu) plus ionizing irradiation (IR). The reprogrammed CAR T cells acquire early memory-like characteristics, potent cytotoxicity, enhanced in vivo expansion, persistence, and decreased exhaustion. Tumors stressed by DSF/Cu and IR also reprogram and reverse the immunosuppressive TME in humanized mice. The reprogrammed CAR T cells, derived from peripheral blood mononuclear cells of healthy donors or metastatic female breast cancer patients, induce robust, sustained memory and curative anti-solid tumor responses in multiple xenograft mouse models, establishing proof of concept for empowering CAR T by stressing tumor as a promising therapy for solid tumors.


Asunto(s)
Neoplasias de la Mama , Receptores Quiméricos de Antígenos , Humanos , Femenino , Animales , Ratones , Leucocitos Mononucleares , Microambiente Tumoral , Neoplasias de la Mama/terapia , Modelos Animales de Enfermedad , Inmunosupresores , Linfocitos T
2.
J Maxillofac Oral Surg ; 22(3): 559-570, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37534349

RESUMEN

Objective: A specific type of mesenchymal stem/progenitor cells (MSPCs), CD105+ is reported to aid in cartilage regeneration through TGF-ß/Smad2-signalling. The purpose of this study was to identify and characterize CD105+ MSPCs in temporomandibular joint (TMJ) cartilage. Materials and Methods: MSPCs were isolated from mouse TMJ condyle explants and evaluated for their clonogenicity and pluripotential abilities. MSPC were examined for CD105 antigen using immunohistochemistry and flow cytometry. Results: Immunohistochemistry revealed presence of CD105+ MSPCs in the proliferative zone of condyle's cartilage. Only 0.2% of isolated MSPCs exhibited CD105, along with the stem cell surface markers CD44 and Sca-1. In CD105+ MSPCs, intracellular immunostaining revealed significantly higher (p < 0.05) protein levels of collagen type 1, 2, proteoglycan 4. Ability for chondrogenic differentiation was found to be significantly higher (p < 0.05) after 4 weeks compared to CD105- cells, using alcian blue staining. CD105+ cells were found to resemble an early MSPC subgroup with significantly higher gene expression of biglycan, proteoglycan 4, collagen type 2, Gli2, Sox5 (p < 0.001) and Sox9 (p < 0.05). In contrast, significantly lower levels of Runx2 (p < 0.05), Osterix, Trps1, Col10a1 (p < 0.01), Ihh (p < 0.001) related to chondrocyte senescence and commitment to osteogenic lineage, were observed compared to CD105- cells. Conclusion: The study showed the existence of a CD105+ MSPC subgroup within TMJ fibrocartilage that may be activated to aid in fibrocartilage repair.

3.
J Stomatol Oral Maxillofac Surg ; 124(6): 101479, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37080358

RESUMEN

Recent studies have shown that bisphosphonates can also impact osteoblasts besides osteoclasts. This study aimed to evaluate the effects of different concentrations of Zoledronic acid (ZA) during the osteogenic differentiation of human Bone Marrow Stem Cells (hBMSCs) in vitro. Thus, osteogenic differentiation of hBMSCs was conducted with different concentrations of Zoledronic Acid (ZA) (0, 0.1, 1.0, and 5.0 µM) for the first 3 days. Cell metabolism was quantified at 1-, 3-, 7-, and 14 days. At 7- and 14-days, the following analyses were performed: 1) mineralization nodule assay, 2) LIVE/DEAD™, 3) cell adhesion and spreading, 4) alkaline phosphatase (ALP) activity, and 5) qPCR analysis for RUNX-2), ALPL, and COL1 A1. Data were analyzed by ANOVA 2-way, followed by Tukey's post hoc test (p < 0.05). Cell metabolism (3-, 7-, and 14-days) (p < 0.001), mineralization (7-, 14-days) (p < 0.001), and ALP activity (14-days) (p < 0.001) were reduced in ZA 5.0 µM when compared to control (no ZA). Also, ZA 5.0 µM downregulated the expression of RUNX2 at 7- and 14-days (p < 0.001). It is possible to conclude that ZA (5.0 µM) can impair hBMSC differentiation into osteoblasts and interferes with its mineralization phase.


Asunto(s)
Difosfonatos , Osteogénesis , Humanos , Ácido Zoledrónico/farmacología , Difosfonatos/farmacología , Diferenciación Celular , Células de la Médula Ósea
4.
Res Sq ; 2023 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-36865255

RESUMEN

The poor efficacy of chimeric antigen receptor T-cell therapy (CAR T) for solid tumor is due to insufficient CAR T cell tumor infiltration, in vivo expansion, persistence, and effector function, as well as exhaustion, intrinsic target antigen heterogeneity or antigen loss of target cancer cells, and immunosuppressive tumor microenvironment (TME). Here we describe a broadly applicable nongenetic approach that simultaneously addresses the multiple challenges of CAR T as a therapy for solid tumors. The approach massively reprograms CAR T cells by exposing them to stressed target cancer cells which have been exposed to the cell stress inducer disulfiram (DSF) and copper (Cu)(DSF/Cu) plus ionizing irradiation (IR). The reprogrammed CAR T cells acquired early memory-like characteristics, potent cytotoxicity, enhanced in vivo expansion, persistence, and decreased exhaustion. Tumors stressed by DSF/Cu and IR also reprogrammed and reversed immunosuppressive TME in humanized mice. The reprogrammed CAR T cells, derived from peripheral blood mononuclear cells (PBMC) of healthy or metastatic breast cancer patients, induced robust, sustained memory and curative anti-solid tumor responses in multiple xenograft mouse models, establishing proof of concept for empowering CAR T by stressing tumor as a novel therapy for solid tumor.

5.
J Stomatol Oral Maxillofac Surg ; 124(1S): 101373, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36584767

RESUMEN

BACKGROUND: Tissue engineering of skin and mucosa is essential for the esthetic and functional reconstruction of individuals disfigured by trauma, resection surgery, or severe burns while overcoming the limited amount of autograft and donor site morbidity. PURPOSE: We aimed to determine whether a combination of Gelatin-methacryloyl (GelMA) hydrogel scaffold alone or loaded with either dental pulp stem cells (DPSCs) and/or vascular endothelial growth factor (VEGF) could improve skin wound healing in rats. MATERIALS AND METHODS: Four 10 mm full-thickness skin defects were created on the dorsum of 15 Sprague-Dawley rats. The wounds were treated with GelMA alone, GelMA+DPSCs, or GelMA+DPSCs+VEGF. Unprotected wounds were used as controls. Animals were euthanized at 1-, 2-, and 4 weeks post-surgery, and the healing wounds were harvested for clinical, histological, and RT-PCR analysis. RESULTS: No signs of clinical inflammation were observed among all groups. Few and sparse mononuclear inflammatory cells were observed in GelMA+DPSCs and GelMA+DPSCs+VEGF groups at 2 weeks, with complete epithelialization of the wounds. At 4 weeks, the epidermis in GelMA+DPSCs and GelMA+DPSCs+VEGF groups was indistinguishable from the empty defect and GelMA groups. The decrease in cellularity and increase in density of collagen fibers were observed over time in both GelMA+DPSCs and GelMA+DPSCs+VEGF groups but were more evident in the GelMA+DPSCs+VEGF group. The GelMA+DPSCs+VEGF group showed a higher expression of the KER 10 gene at all time points compared with the other groups. Expression of Col1 A1 and TGFß-1 were not statistically different over time neither among the groups. CONCLUSION: GelMA scaffolds loaded with DPSCs, and VEGF accelerated the re-epithelialization of skin wounds.


Asunto(s)
Gelatina , Factor A de Crecimiento Endotelial Vascular , Ratas , Animales , Humanos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ratas Sprague-Dawley , Pulpa Dental/metabolismo , Células Madre
6.
Artículo en Inglés | MEDLINE | ID: mdl-36241596

RESUMEN

OBJECTIVES: Mesenchymal stem/progenitor cells (MSPCs) are critical for tissue regeneration. Moreover, the CD105 antigen identifies early MSPCs with increased chondrogenic differentiation ability. We hypothesized that amine-(NH2)-functionalized biosilica incorporating hydrogel scaffolds, seeded with mCoSPCs105+ would contribute to creating tissue-engineered scaffolds, capable of de novo cartilage synthesis. MATERIALS AND METHODS: Scaffolds were characterized by water uptake, lysozyme degradation, axial compression, scanning electron microscopy, and energy-dispersive X-ray spectroscopy. Differentiation stimulus of scaffold functionalization was evaluated using Alcian blue staining. Cartilage-forming abilities of mCoSPCs105+ were evaluated using Quantitative reverse transcription polymerase chain reaction (qRT-PCR) and immunohistochemistry. RESULTS: Biosilica particle incorporation into scaffolds resulted in increased water uptake capacity and compression force withstanding abilities. Amine-(NH2)-group functionalization of biosilica led to significantly increased stem cell differentiation potential, by Alcian blue staining, in the first 3 weeks. Scaffold attachment and viable cell proliferation were observed for 6 weeks under chondrogenic differentiation. Downregulation of Runx2, an increase of Col10a1, Ihh, and maintenance of Sox9, was seen under these culture conditions. mCoSPCs105+ gene expression pattern was defined by the significant upregulation of Col1a1, Col2a1, Prg4, and Agc-1 over 6 weeks of incubation compared to the unsorted control. Immunostaining of cell-seeded scaffolds revealed significantly higher secretion of proteins relevant to cartilage extracellular matrix. CONCLUSION: The preselecting of CD105+ phenotype in MSPCs may enhance tissue regeneration of fibrocartilage and biosilica nanoparticles may be a beneficial additive in tissue engineering of scaffolds.


Asunto(s)
Hidrogeles , Células Madre Mesenquimatosas , Ratones , Animales , Hidrogeles/química , Hidrogeles/metabolismo , Azul Alcián/metabolismo , Diferenciación Celular , Andamios del Tejido/química , Ingeniería de Tejidos , Células Madre Mesenquimatosas/metabolismo , Condrogénesis , Células Cultivadas
7.
Nature ; 604(7904): 190-194, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35355020

RESUMEN

Type A γ-aminobutyric acid receptors (GABAARs) are pentameric ligand-gated chloride channels that mediate fast inhibitory signalling in neural circuits1,2 and can be modulated by essential medicines including general anaesthetics and benzodiazepines3. Human GABAAR subunits are encoded by 19 paralogous genes that can, in theory, give rise to 495,235 receptor types. However, the principles that govern the formation of pentamers, the permutational landscape of receptors that may emerge from a subunit set and the effect that this has on GABAergic signalling remain largely unknown. Here we use cryogenic electron microscopy to determine the structures of extrasynaptic GABAARs assembled from α4, ß3 and δ subunits, and their counterparts incorporating γ2 instead of δ subunits. In each case, we identified two receptor subtypes with distinct stoichiometries and arrangements, all four differing from those previously observed for synaptic, α1-containing receptors4-7. This, in turn, affects receptor responses to physiological and synthetic modulators by creating or eliminating ligand-binding sites at subunit interfaces. We provide structural and functional evidence that selected GABAAR arrangements can act as coincidence detectors, simultaneously responding to two neurotransmitters: GABA and histamine. Using assembly simulations and single-cell RNA sequencing data8,9, we calculated the upper bounds for receptor diversity in recombinant systems and in vivo. We propose that differential assembly is a pervasive mechanism for regulating the physiology and pharmacology of GABAARs.


Asunto(s)
Benzodiazepinas , Receptores de GABA-A , Transducción de Señal , Benzodiazepinas/farmacología , Sitios de Unión , Microscopía por Crioelectrón , Histamina/metabolismo , Humanos , Ligandos , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , RNA-Seq , Receptores de GABA-A/química , Receptores de GABA-A/metabolismo , Receptores de GABA-A/ultraestructura , Análisis de la Célula Individual , Ácido gamma-Aminobutírico/metabolismo
8.
Eur J Med Chem ; 194: 112261, 2020 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-32247113

RESUMEN

The pentameric γ-aminobutyric acid type A receptors (GABAARs) are the major inhibitory ligand-gated ion channels in the central nervous system. They mediate diverse physiological functions, mutations in them are associated with mental disorders and they are the target of many drugs such as general anesthetics, anxiolytics and anti-convulsants. The five subunits of synaptic GABAARs are arranged around a central pore in the order ß-α-ß-α-γ. In the outer third of the transmembrane domain (TMD) drugs may bind to five homologous intersubunit binding sites. Etomidate binds between the pair of ß - α subunit interfaces (designated as ß+/α-) and R-mTFD-MPAB binds to an α+/ß- and an γ+/ß- subunit interface (a ß- selective ligand). Ligands that bind selectively to other homologous sites have not been characterized. We have synthesized a novel photolabel, (2,6-diisopropyl-4-(3-(trifluoromethyl)-3H-diazirin-3-yl)phenyl)methanol or pTFD-di-iPr-BnOH). It is a potent general anesthetic that positively modulates agonist and benzodiazepine binding. It enhances GABA-induced currents, shifting the GABA concentration-response curve to lower concentrations. Photolabeling-protection studies show that it has negligible affinity for the etomidate sites and high affinity for only one of the two R-mTFD-MPAB sites. Exploratory site-directed mutagenesis studies confirm the latter conclusions and hint that pTFD-di-iPr-BnOH may bind between the α+/ß- and α+/γ- subunits in the TMD, making it an α+ ligand. The latter α+/γ- site has not previously been implicated in ligand binding. Thus, pTFD-di-iPr-BnOH is a promising new photolabel that may open up a new pharmacology for synaptic GABAARs.


Asunto(s)
Anestésicos Generales/farmacología , Propofol/farmacología , Receptores de GABA-A/metabolismo , Regulación Alostérica/efectos de los fármacos , Anestésicos Generales/síntesis química , Anestésicos Generales/química , Animales , Sitios de Unión/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Modelos Moleculares , Estructura Molecular , Procesos Fotoquímicos , Propofol/síntesis química , Propofol/química , Relación Estructura-Actividad , Xenopus
9.
Anesthesiology ; 131(6): 1276-1291, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31567362

RESUMEN

BACKGROUND: Transgenic mouse studies suggest that γ-aminobutyric acid type A (GABAA) receptors containing ß3 subunits mediate important effects of etomidate, propofol, and pentobarbital. Zebrafish, recently introduced for rapid discovery and characterization of sedative-hypnotics, could also accelerate pharmacogenetic studies if their transgenic phenotypes reflect those of mammals. The authors hypothesized that, relative to wild-type, GABAA-ß3 functional knock-out (ß3) zebrafish would show anesthetic sensitivity changes similar to those of ß3 mice. METHODS: Clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 mutagenesis was used to create a ß3 zebrafish line. Wild-type and ß3 zebrafish were compared for fertility, growth, and craniofacial development. Sedative and hypnotic effects of etomidate, propofol, pentobarbital, alphaxalone, ketamine, tricaine, dexmedetomidine, butanol, and ethanol, along with overall activity and thigmotaxis were quantified in 7-day postfertilization larvae using video motion analysis of up to 96 animals simultaneously. RESULTS: Xenopus oocyte electrophysiology showed that the wild-type zebrafish ß3 gene encodes ion channels activated by propofol and etomidate, while the ß3 zebrafish transgene does not. Compared to wild-type, ß3 zebrafish showed similar morphology and growth, but more rapid swimming. Hypnotic EC50s (mean [95% CI]) were significantly higher for ß3 versus wild-type larvae with etomidate (1.3 [1.0 to 1.6] vs. 0.6 [0.5 to 0.7] µM; P < 0.0001), propofol (1.1 [1.0 to 1.4] vs. 0.7 [0.6 to 0.8] µM; P = 0.0005), and pentobarbital (220 [190 to 240] vs. 130 [94 to 179] µM; P = 0.0009), but lower with ethanol (150 [106 to 213] vs. 380 [340 to 420] mM; P < 0.0001) and equivalent with other tested drugs. Comparing ß3 versus wild-type sedative EC50s revealed a pattern similar to hypnosis. CONCLUSIONS: Global ß3 zebrafish are selectively insensitive to the same few sedative-hypnotics previously reported in ß3 transgenic mice, indicating phylogenetic conservation of ß3-containing GABAA receptors as anesthetic targets. Transgenic zebrafish are potentially valuable models for sedative-hypnotic mechanisms research.


Asunto(s)
Anestésicos/administración & dosificación , Hipnóticos y Sedantes/administración & dosificación , Locomoción/efectos de los fármacos , Locomoción/genética , Receptores de GABA-A/deficiencia , Receptores de GABA-A/genética , Animales , Animales Modificados Genéticamente , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Femenino , Ratones , Ratones Noqueados , Ratones Transgénicos , Xenopus laevis , Pez Cebra
10.
J Pharmacol Exp Ther ; 368(1): 100-105, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30389723

RESUMEN

Central α4ßδ receptors are the most abundant isoform of δ subunit-containing extrasynaptic GABAA receptors that mediate tonic inhibition. Although the amplitude of GABA-activated currents through α4ßδ receptors is modulated by multiple general anesthetics, the effects of general anesthetics on desensitization and deactivation of α4ßδ receptors remain unknown. In the current study, we investigated the effect of etomidate, a potent general anesthetic, on the kinetics and the pseudo steady-state current amplitude of α4ß3δ receptors inducibly expressed in human embryonic kidney 293 TetR cells. Etomidate directly activates α4ß3δ receptors in a concentration-dependent manner. Etomidate at a clinically relevant concentration (3.2 µM) enhances maximal response without altering the EC50 of GABA concentration response. Etomidate also increases the extent of desensitization and prolongs the deactivation of α4ß3δ receptors in the presence of maximally activating concentrations of GABA (1 mM). To mimic the modulatory effect of etomidate on tonic currents, long pulses (30-60 seconds) of a low GABA concentration (1 µM) were applied to activate α4ß3δ receptors in the absence and presence of etomidate. Although etomidate increases the desensitization of α4ß3δ receptors, the pseudo steady-state current amplitude at 1 µM GABA is augmented by etomidate. Our data demonstrate that etomidate enhances the pseudo steady-state current of α4ß3δ receptors evoked by a GABA concentration comparable to an ambient GABA level, suggesting that α4ß3δ receptors may mediate etomidate's anesthetic effect in the brain.


Asunto(s)
Etomidato/farmacología , Agonistas del GABA/farmacología , Hipnóticos y Sedantes/farmacología , Receptores de GABA-A/biosíntesis , Relación Dosis-Respuesta a Droga , Expresión Génica , Células HEK293 , Humanos , Receptores de GABA-A/genética , Ácido gamma-Aminobutírico/farmacología
11.
Anesthesiology ; 129(3): 459-476, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29894316

RESUMEN

WHAT WE ALREADY KNOW ABOUT THIS TOPIC: WHAT THIS ARTICLE TELLS US THAT IS NEW: BACKGROUND:: Many general anesthetics were discovered empirically, but primary screens to find new sedative-hypnotics in drug libraries have not used animals, limiting the types of drugs discovered. The authors hypothesized that a sedative-hypnotic screening approach using zebrafish larvae responses to sensory stimuli would perform comparably to standard assays, and efficiently identify new active compounds. METHODS: The authors developed a binary outcome photomotor response assay for zebrafish larvae using a computerized system that tracked individual motions of up to 96 animals simultaneously. The assay was validated against tadpole loss of righting reflexes, using sedative-hypnotics of widely varying potencies that affect various molecular targets. A total of 374 representative compounds from a larger library were screened in zebrafish larvae for hypnotic activity at 10 µM. Molecular mechanisms of hits were explored in anesthetic-sensitive ion channels using electrophysiology, or in zebrafish using a specific reversal agent. RESULTS: Zebrafish larvae assays required far less drug, time, and effort than tadpoles. In validation experiments, zebrafish and tadpole screening for hypnotic activity agreed 100% (n = 11; P = 0.002), and potencies were very similar (Pearson correlation, r > 0.999). Two reversible and potent sedative-hypnotics were discovered in the library subset. CMLD003237 (EC50, ~11 µM) weakly modulated γ-aminobutyric acid type A receptors and inhibited neuronal nicotinic receptors. CMLD006025 (EC50, ~13 µM) inhibited both N-methyl-D-aspartate and neuronal nicotinic receptors. CONCLUSIONS: Photomotor response assays in zebrafish larvae are a mechanism-independent platform for high-throughput screening to identify novel sedative-hypnotics. The variety of chemotypes producing hypnosis is likely much larger than currently known.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Hipnóticos y Sedantes/farmacología , Larva/efectos de los fármacos , Locomoción/efectos de los fármacos , Reflejo de Enderezamiento/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Femenino , Larva/fisiología , Locomoción/fisiología , Masculino , Ratas , Ratas Sprague-Dawley , Reflejo de Enderezamiento/fisiología , Xenopus , Pez Cebra
12.
PLoS One ; 13(1): e0191583, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29352320

RESUMEN

The inhibitory γ-aminobutyric acid type A receptors are implicated in numerous physiological processes, including cognition and inhibition of neurotransmission, rendering them important molecular targets for many classes of drugs. Functionally, the entire GABAAR family of receptors can be subdivided into phasic, fast acting synaptic receptors, composed of α-, ß- and γ-subunits, and tonic extrasynaptic receptors, many of which contain the δ-subunit in addition to α- and ß-subunits. Whereas the subunit arrangement of the former group is agreed upon, that of the αßδ GABAARs remains unresolved by electrophysiological and pharmacological research. To resolve such issues will require biophysical techniques that demand quantities of receptor that have been previously unavailable. Therefore, we have engineered a stable cell line with tetracycline inducible expression of human α4-, ß3- and N-terminally Flag-tagged δ-subunits. This cell line achieved a specific activity between 15 and 20 pmol [3H]muscimol sites/mg of membrane protein, making it possible to obtain 1 nmole of purified α4ß3δ GABAAR from sixty 15-cm culture dishes. When induced, these cells exhibited agonist-induced currents with characteristics comparable to those previously reported for this receptor and a pharmacology that included strong modulation by etomidate and the δ-subunit-specific ligand, DS2. Immunoaffinity purification and reconstitution in CHAPS/asolectin micelles resulted in the retention of equilibrium allosteric interactions between the separate agonist, anesthetic and DS2 sites. Moreover, all three subunits retained glycosylation. The establishment of this well-characterized cell line will allow molecular level studies of tonic receptors to be undertaken.


Asunto(s)
Receptores de GABA-A/biosíntesis , Fenómenos Electrofisiológicos , Células HEK293 , Humanos , Cinética , Ingeniería de Proteínas , Subunidades de Proteína , Ensayo de Unión Radioligante , Receptores de GABA-A/genética , Receptores de GABA-A/aislamiento & purificación , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Transfección , Ácido gamma-Aminobutírico/metabolismo
13.
Cancer Res ; 77(21): 5938-5951, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28916655

RESUMEN

IL2 is an immunostimulatory cytokine for key immune cells including T cells and natural killer (NK) cells. Systemic IL2 supplementation could enhance NK-mediated immunity in a variety of diseases ranging from neoplasms to viral infection. However, its systemic use is restricted by its serious side effects and limited efficacy due to activation of T regulatory cells (Tregs). IL2 signaling is mediated through interactions with a multi-subunit receptor complex containing IL2Rα, IL2Rß, and IL2Rγ. Adult natural killer (NK) cells express only IL2Rß and IL2Rγ subunits and are therefore relatively insensitive to IL2. To overcome these limitations, we created a novel chimeric IL2-IL2Rß fusion protein of IL2 and its receptor IL2Rß joined via a peptide linker (CIRB). NK92 cells expressing CIRB (NK92CIRB) were highly activated and expanded indefinitely without exogenous IL2. When compared with an IL2-secreting NK92 cell line, NK92CIRB were more activated, cytotoxic, and resistant to growth inhibition. Direct contact with cancer cells enhanced the cytotoxic character of NK92CIRB cells, which displayed superior in vivo antitumor effects in mice. Overall, our results showed how tethering IL2 to its receptor IL2Rß eliminates the need for IL2Rα and IL2Rß, offering a new tool to selectively activate and empower immune therapy. Cancer Res; 77(21); 5938-51. ©2017 AACR.


Asunto(s)
Citotoxicidad Inmunológica/inmunología , Subunidad beta del Receptor de Interleucina-2/inmunología , Interleucina-2/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Animales , División Celular/inmunología , Línea Celular , Línea Celular Tumoral , Técnicas de Cocultivo , Células HEK293 , Células Hep G2 , Humanos , Inmunoterapia Adoptiva/métodos , Interleucina-2/genética , Interleucina-2/metabolismo , Subunidad beta del Receptor de Interleucina-2/genética , Subunidad beta del Receptor de Interleucina-2/metabolismo , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/trasplante , Masculino , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/terapia , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
14.
J Biol Chem ; 291(51): 26529-26539, 2016 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-27821594

RESUMEN

Extrasynaptic γ-aminobutyric acid type A receptors (GABAARs),which contribute generalized inhibitory tone to the mammalian brain, are major targets for general anesthetics. To identify anesthetic binding sites in an extrasynaptic GABAAR, we photolabeled human α4ß3δ GABAARs purified in detergent with [3H]azietomidate and a barbiturate, [3H]R-mTFD-MPAB, photoreactive anesthetics that bind with high selectivity to distinct but homologous intersubunit binding sites in the transmembrane domain of synaptic α1ß3γ2 GABAARs. Based upon 3H incorporation into receptor subunits resolved by SDS-PAGE, there was etomidate-inhibitable labeling by [3H]azietomidate in the α4 and ß3 subunits and barbiturate-inhibitable labeling by [3H]R-mTFD-MPAB in the ß3 subunit. These sites did not bind the anesthetic steroid alphaxalone, which enhanced photolabeling, or DS-2, a δ subunit-selective positive allosteric modulator, which neither enhanced nor inhibited photolabeling. The amino acids labeled by [3H]azietomidate or [3H]R-mTFD-MPAB were identified by N-terminal sequencing of fragments isolated by HPLC fractionation of enzymatically digested subunits. No evidence was found for a δ subunit contribution to an anesthetic binding site. [3H]azietomidate photolabeling of ß3Met-286 in ßM3 and α4Met-269 in αM1 that was inhibited by etomidate but not by R-mTFD-MPAB established that etomidate binds to a site at the ß3+-α4- interface equivalent to its site in α1ß3γ2 GABAARs. [3H]Azietomidate and [3H]R-mTFD-MPAB photolabeling of ß3Met-227 in ßM1 established that these anesthetics also bind to a homologous site, most likely at the ß3+-ß3- interface, which suggests a subunit arrangement of ß3α4ß3δß3.


Asunto(s)
Barbitúricos/química , Receptores de GABA/química , Barbitúricos/metabolismo , Sitios de Unión , Células HEK293 , Humanos , Estructura Cuaternaria de Proteína , Receptores de GABA/metabolismo
15.
Oncol Rep ; 26(1): 281-6, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21503586

RESUMEN

Malignant gliomas are aggressive and angiogenic tumors with high VEGF content. Consequently, approaches such as metronomic chemotherapy, which have an anti-angiogenic effect, are being investigated. However, a lack of an appropriate technique that can facilitate the identification of vascular changes during antiangiogenic treatments has restricted therapeutic optimization. We have investigated the potential of tumor pO2 as a marker to detect vascular changes during metronomic chemotherapy. Electron paramagnetic resonance (EPR) oximetry was used to repeatedly assess tumor pO2 during metronomic cyclophosphamide treatment of subcutaneous 9L tumors. The 9L tumors were hypoxic with a pO2 of 5.6-8 mmHg and a tumor volume of 247-300 mm3 prior to any treatment. Tumor pO2 increased significantly to 19.7 mmHg on day 10 and remained at an elevated level until day 33 during 4 weekly treatments with 140 mg/kg cyclophosphamide. A significant decrease in the tumor volume on days 21-31 occurred in the cyclophosphamide group, while the tumor volume of the control group significantly increased during measurements for two weeks. A significant tumor growth delay was achieved with two weekly treatments of cyclophosphamide plus radiotherapy (4 Gy x 5) as compared to control, cyclophosphamide and radiotherapy alone groups. The results indicate the potential of EPR oximetry to assess tumor pO2 during metronomic chemotherapy. The ability to identify the duration of an increase in tumor pO2, therapeutic window, non-invasively by EPR oximetry could have a significant impact on the optimization of antiangiogenic approaches for the treatment of gliomas. This vital information could also be used to schedule radiotherapy to enhance therapeutic outcome.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Ciclofosfamida/farmacología , Glioma/tratamiento farmacológico , Glioma/metabolismo , Oximetría/métodos , Animales , Antineoplásicos Alquilantes/farmacología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Espectroscopía de Resonancia por Spin del Electrón , Humanos , Ratones , Ratones SCID , Oxígeno/química , Oxígeno/metabolismo , Ratas , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
Hum Gene Ther ; 19(12): 1383-400, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18771358

RESUMEN

We constructed an oncolytic adenovirus, Adeno-hTERT-E1A, with deletions of the viral E1B, E3A, and E3B regions and insertion of a human telomerase reverse transcriptase (hTERT) promoter-driven early viral 1A (E1A) cassette that confers high transcriptional activity in multiple human tumor cell lines. The oncolytic potential of Adeno-hTERT-E1A was characterized in comparison with that of the E1B-55 kDa- and E3B-region-deleted oncolytic adenovirus ONYX-015. Tumor cells infected with Adeno-hTERT-E1A expressed dramatically higher levels of E1A oncoprotein, underwent enhanced lysis, and displayed an earlier and higher apoptotic index than cells infected with ONYX-015. Despite the increase in virus-induced apoptotic death, Adeno-hTERT-E1A replicated and produced functional progeny leading to viral spread, but with reduced efficiency compared with ONYX-015, in particular in A549 cells. Virus-induced E1A expression, host cell apoptosis, viral hexon protein production, and DNA synthesis were markedly reduced in primary human hepatocytes after infection with Adeno-hTERT-E1A as compared with ONYX-015. The strong oncolytic activity of Adeno-hTERT-E1A in tumor cell culture translated into superior antitumor activity in vivo in an MDA-MB-231 solid tumor xenograft model. Adeno-hTERT-E1A thus has strong therapeutic potential and an improved safety profile compared with ONYX-015, which may lead to reduced toxicity in the clinic.


Asunto(s)
Adenoviridae/genética , Proteínas E1B de Adenovirus/genética , Eliminación de Gen , Viroterapia Oncolítica , Regiones Promotoras Genéticas , Telomerasa/genética , Secuencia de Bases , Línea Celular , Cartilla de ADN , Humanos , Reacción en Cadena de la Polimerasa
17.
Curr Cancer Drug Targets ; 7(3): 285-301, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17504125

RESUMEN

Replication-conditional, oncolytic adenoviruses are emerging as powerful tools in the warfare on cancer. The ability to modify cell-specific infectivity or tissue-specific replication machinery, as well as the possibility of modifying viral-cellular protein interactions with cellular checkpoint regulators are emerging as new trends in the design of safer and more effective adenoviruses. The integration of oncolytic adenoviruses with mainstream cancer therapies, such as chemotherapy and radiotherapy, continues to yield significant therapeutic benefits. Adenoviruses can be armed with prodrug-activating enzymes as well as tumor suppressor genes or anti-angiogenic factors, thus providing for enhanced anti-tumor therapy and reduced host toxicity. Thus far, encouraging results have been obtained from extensive preclinical and human clinical studies. However, there is a need to improve adenoviral vectors to overcome unresolved problems facing this promising anti-cancer agent, chief among these issues is the adenovirus-triggered immune response threatening its efficacy. The continued expansion of the knowledge base of adenovirus biology will likely lead to further improvements in the design of the ideal oncolytic adenoviruses for cancer treatment.


Asunto(s)
Adenovirus Humanos/fisiología , Neoplasias/tratamiento farmacológico , Neoplasias/virología , Replicación Viral/fisiología , Adenovirus Humanos/genética , Animales , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Replicación Viral/efectos de los fármacos , Replicación Viral/genética
18.
Mol Cancer Ther ; 5(3): 541-55, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16546968

RESUMEN

Gene therapy using the prodrug-activating enzyme P450 2B6 has shown substantial promise in preclinical and initial clinical studies with the P450 prodrugs cyclophosphamide and ifosfamide. We sought to optimize this therapy using the canine P450 enzyme 2B11, which activates cyclophosphamide and ifosfamide with Km of 80 to 160 micromol/L, approximately 10- to 20-fold lower than the Km of P450 2B6. Retrovirus encoding a P450 2B11-internal ribosome entry signal-P450 reductase expression cassette induced marked cyclophosphamide and ifosfamide cytotoxicity toward 9L gliosarcoma cells and exhibited an impressive bystander killing effect at micromolar prodrug concentrations, where P450 2B6 displayed low activity. Adeno-2B11, a replication-defective, E1/E3 region-deleted adenovirus engineered to coexpress P450 2B11 and P450 reductase, dramatically increased tumor cell-catalyzed cyclophosphamide 4-hydroxylation and cytotoxicity compared with Adeno-2B6 and effected strong bystander killing at low (20 micromol/L) cyclophosphamide concentrations. Further increases in cyclophosphamide cytotoxicity were obtained in several human cancer cell lines, including a 4-hydroperoxycyclophosphamide-resistant MCF-7 breast cancer cell line, when Adeno-2B11 was combined with Onyx-017, an E1b-55-kDa gene-deleted, tumor cell-replicating adenovirus that coamplifies and facilitates tumor cell spread of Adeno-2B11. To evaluate the therapeutic effect of P450 2B11 expression in vivo, 9L gliosarcoma cells transduced with P450-expressing retrovirus were grown as solid s.c. tumors in immunodeficient mice. Cyclophosphamide treatment on a metronomic, 6-day repeating schedule led to full regression of 9L/2B11 tumors but not P450-deficient control tumors, resulting in a tumor-free period lasting up to approximately 100 days. 9L/2B6 tumors regressed more slowly and exhibited a tumor-free period of only 21 to 39 days. Thus, P450 gene-directed enzyme prodrug therapy can be greatly improved by using the low Km P450 enzyme 2B11, which catalyzes intratumoral activation of cyclophosphamide and ifosfamide at pharmacologically relevant drug concentrations.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Hidrocarburo de Aril Hidroxilasas/genética , Ciclofosfamida/uso terapéutico , Terapia Genética/métodos , Neoplasias/terapia , Profármacos/uso terapéutico , Esteroide Hidroxilasas/genética , Adenoviridae/genética , Animales , Antineoplásicos Alquilantes/metabolismo , Catálisis , Terapia Combinada , Ciclofosfamida/metabolismo , Familia 2 del Citocromo P450 , Resistencia a Antineoplásicos/genética , Vectores Genéticos , Humanos , Ifosfamida/metabolismo , Ifosfamida/uso terapéutico , Neoplasias/tratamiento farmacológico , Profármacos/metabolismo , Ratas , Retroviridae/genética , Transfección , Células Tumorales Cultivadas
19.
Drug Metab Dispos ; 33(8): 1083-96, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16049130

RESUMEN

Drug-metabolizing enzymes and drug transporters are key determinants of the pharmacokinetics and pharmacodynamics of many antineoplastic agents. Metabolism and transport influence the cytotoxic effects of antineoplastic agents in target tumor cells and normal host tissues. This article summarizes several state-of-the-art approaches to enhancing the effectiveness and safety of cancer therapy based on recent developments in our understanding of antineoplastic drug metabolism and transport. Advances in four interrelated research areas presented at a recent symposium sponsored by the Division for Drug Metabolism of the American Society for Pharmacology and Experimental Therapeutics (Experimental Biology 2004; Washington D.C., April 17-21, 2004) are discussed: 1) interactions of anthracyclines with drug-metabolizing enzymes; 2) use of hypoxia-selective gene-directed enzyme prodrug therapy (GDEPT) in combination with bioreductive prodrugs; 3) synergy between glutathione conjugation and conjugate efflux in conferring resistance to electrophilic toxins; and 4) use of cytochromes P450 as prodrug-activating enzymes in GDEPT strategies. A clear theme emerged from this symposium: drug metabolism and transport processes can be modulated and exploited in ways that may offer distinct therapeutic advantages in the management of patients with cancer.


Asunto(s)
Antineoplásicos/metabolismo , Antineoplásicos/uso terapéutico , Sistema Enzimático del Citocromo P-450/metabolismo , Terapia Genética , Hígado/metabolismo , Neoplasias/tratamiento farmacológico , Profármacos/metabolismo , Profármacos/uso terapéutico , Animales , Antibióticos Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/farmacología , Transporte Biológico , Línea Celular Tumoral/efectos de los fármacos , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/uso terapéutico , Doxorrubicina/metabolismo , Resistencia a Antineoplásicos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/prevención & control , Vectores Genéticos , Glutatión/metabolismo , Glutatión Transferasa/antagonistas & inhibidores , Glutatión Transferasa/metabolismo , Humanos , Hígado/enzimología , Neoplasias/metabolismo , Oxidación-Reducción , Profármacos/clasificación
20.
Drug Metab Dispos ; 33(9): 1261-7, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15919850

RESUMEN

The anticancer prodrug ifosfamide (IFA) contains a chiral phosphorous atom and is administered in the clinic as a racemic mixture of R-IFA and S-IFA. Hepatic cytochrome P450 (P450) enzymes exhibit enantioselective preferences in the metabolism of R-IFA and S-IFA; however, the impact of this selectivity on P450-dependent anticancer activity is not known. Presently, the metabolism and cytotoxicity of R-IFA and S-IFA were determined in 9L gliosarcoma and Chinese hamster ovary tumor cells expressing an IFA-activating P450 enzyme and by in vitro steady-state kinetic analysis using cDNA-expressed P450 enzymes. Tumor cells expressing P450 enzyme CYP3A4 were the most sensitive to R-IFA cytotoxicity, whereas tumor cells expressing CYP2B1 or CYP2B6 were most sensitive to cyclophosphamide (CPA), an isomer of IFA. Correspondingly, CYP3A4-expressing cells and cDNA-expressed CYP3A4 metabolized R-IFA to yield the active, 4-hydroxylated metabolite at a 2- to 3-fold higher rate than they metabolized S-IFA or CPA. CYP2B cells and cDNA-expressed CYP2B enzymes metabolized CPA almost exclusively by 4-hydroxylation, whereas R-IFA and S-IFA were substantially converted to inactive, N-dechloroethylated metabolites. Further investigation revealed that CYP3A1, a rat enzyme, exhibited superior kinetic properties compared with the human enzyme CYP3A4, with R-IFA and S-IFA both metabolized with high catalytic efficiency by 4-hydroxylation and with a K(m) value of 200 microM, approximately 5-fold lower than CYP3A4. Based on these kinetic parameters and metabolic profiles, R-IFA is expected to exert greater anticancer activity than S-IFA or CPA against tumors that express CYP3A enzymes, whereas tumors expressing CYP2B enzymes may be more sensitive to CPA treatment.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Ciclofosfamida/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , Ifosfamida/farmacología , Profármacos/farmacología , Acetaldehído/análogos & derivados , Acetaldehído/farmacología , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cricetinae , Cricetulus , Sistema Enzimático del Citocromo P-450/biosíntesis , Sistema Enzimático del Citocromo P-450/genética , ADN Complementario/metabolismo , Expresión Génica , Humanos , Ifosfamida/análogos & derivados , NADPH-Ferrihemoproteína Reductasa/biosíntesis , NADPH-Ferrihemoproteína Reductasa/genética , NADPH-Ferrihemoproteína Reductasa/metabolismo , Ratas , Estereoisomerismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA