Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Bioorg Med Chem Lett ; 26(1): 133-9, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26602273

RESUMEN

In recent years, mGlu4 has received great research attention because of the potential benefits of mGlu4 activation in treating numerous brain disorders, such as Parkinson's disease (PD). A specific mGlu4 PET radioligand could be an important tool in understanding the role of mGlu4 in both healthy and disease conditions, and also for the development of new drugs. In this study, we synthesized four new N-(methylthiophenyl)picolinamide derivatives 11-14. Of these ligands, 11 and 14 showed high in vitro binding affinity for mGlu4 with IC50 values of 3.4nM and 3.1nM, respectively, and suitable physicochemical parameters. Compound 11 also showed enhanced metabolic stability and good selectivity to other mGluRs. [(11)C]11 and [(11)C]14 were radiolabeled using the [(11)C]methylation of the thiophenol precursors 20a and 20c with [(11)C]CH3I in 19.0% and 34.8% radiochemical yields (RCY), and their specific activities at the end of synthesis (EOS) were 496±138GBq/µmol (n=6) and 463±263GBq/µmol (n=4), respectively. The PET studies showed that [(11)C]11 accumulated fast into the brain and had higher uptake, slower washout and 25% better contrast than [(11)C]2, indicating improved imaging characteristics as PET radiotracer for mGlu4 compared to [(11)C]2. Therefore, [(11)C]11 will be a useful radioligand to investigate mGlu4 in different biological applications.


Asunto(s)
Ácidos Picolínicos/síntesis química , Ácidos Picolínicos/metabolismo , Tomografía de Emisión de Positrones , Ensayo de Unión Radioligante , Radiofármacos/síntesis química , Radiofármacos/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Tiofenos/síntesis química , Tiofenos/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Ligandos , Masculino , Estructura Molecular , Ácidos Picolínicos/química , Unión Proteica , Radiofármacos/análisis , Radiofármacos/química , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad , Especificidad por Sustrato , Tiofenos/química
2.
J Neuroinflammation ; 12: 217, 2015 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-26597638

RESUMEN

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative motor neuron disorder. Genetic studies have linked mutation of the gene SOD1 to ALS pathology as well as several other pathological processes including modulation of glutamatergic function and inflammatory processes. Since therapeutic approaches for ALS are focused on glutamatergic function, we investigated modulation of glutamate transport based on its receptor function as well as excitotoxicity-induced inflammatory response. METHODS: In vivo positron emission tomography (PET) imaging studies of metabotropic glutamate receptor subtype 5 (mGluR5) using [(18)F]FPEB ([(18)F]3-fluoro-5-(2-pyridylethynyl)benzonitrile) and inflammatory response using [(11)C]PBR28 (peripheral benzodiazepine receptor ligand 28) were done in an early and a late phase of neurodegeneration in four ALS mice expressing SOD1-G93A gene and four control base mice (C57/BL6). Accumulation of [(18)F]FPEB and [(11)C]PBR28 were quantitated in several brain areas and spinal cord to determine degeneration-induced modulation. The studies were completed with immunohistochemical analyses of mGluR5 and inflammatory response. RESULTS: These studies showed enhanced binding potential of [(18)F]FPEB in several brain areas including striatum, hippocampus, and frontal cortex. In the whole brain, the binding potential increased 49 ± 9 % from base mice to ALS-type mice and further enhanced 23 ± 4 % during disease progression. Also, in the spinal cord 6-22 %, enhanced accumulation of [(18)F]FPEB was observed during progression of the disease. The accumulation of [(11)C]PBR28 increased by 110 ± 33 % in the whole brain during progression of the disease indicating significant inflammatory process. [(11)C]PBR28 accumulation enhanced 89-264 % in the spinal cord and 204 % in the lungs. The end point immunohistochemical analyses verified the enhanced mGluR5 expression and inflammation. CONCLUSIONS: These results confirm the role of glutamate and inflammation in ALS-type pathology. These data also support the hypothesis that excessive glutamate may contribute to inflammation in the chronic neurodegenerative processes in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/diagnóstico por imagen , Esclerosis Amiotrófica Lateral/metabolismo , Modelos Animales de Enfermedad , Tomografía de Emisión de Positrones , Receptor del Glutamato Metabotropico 5/biosíntesis , Superóxido Dismutasa/biosíntesis , Esclerosis Amiotrófica Lateral/genética , Animales , Progresión de la Enfermedad , Regulación de la Expresión Génica , Humanos , Inflamación/diagnóstico por imagen , Inflamación/genética , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Tomografía de Emisión de Positrones/métodos , Receptor del Glutamato Metabotropico 5/genética , Superóxido Dismutasa/genética
3.
Cancer Gene Ther ; 30(6): 833-844, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36721067

RESUMEN

Leptomeningeal metastasis is a fatal complication of breast cancer which results when cancer cells seed in the meninges. Currently there is no cure, limiting survival to less than four months. Treatment options are palliative. We studied a replication conditional Herpes simplex virus 1 (HSV1) in this regard and present the therapeutic efficacy of oncolytic HSV1 on different stages of breast cancer leptomeningeal metastases growth, namely the lag, intermediate, and exponential phases. These phases characterized in a murine model represent the early, intermediate, and late stages of leptomeningeal disease in patients. In this model, virus was introduced into the ventricular system by stereotactic surgery, the same path cancer cells were introduced to create leptomeningeal metastases. Tumor growth was measured with Gd-MRI and virus replication was assessed by FHBG-PET and Fluc bioluminescence. Imaging results were correlated with H&E and HSV-TK immunohistochemical staining. A remarkable growth inhibition was observed when the lag phase was targeted which was associated with multiple virus replication cycles. The onset of debilitating symptoms was delayed, and survival was lengthened by nearly 2 weeks. A growth inhibition similar to the lag phase was observed when the intermediate phase was targeted, associated with robust virus replication. The regression of existing tumor led to a reversal of neurological symptoms, extending survival by nearly one week. A modest response was observed when the lag phase was targeted lengthening survival by 3 days. Oncolytic HSV1 presents a novel treatment option for breast cancer leptomeningeal metastases with potential for targeting different disease stages where virus replication and tumor response can be monitored with molecular imaging techniques that are in the clinic.


Asunto(s)
Neoplasias de la Mama , Herpesvirus Humano 1 , Ratones , Humanos , Animales , Femenino , Herpesvirus Humano 1/genética , Neoplasias de la Mama/terapia , Neoplasias de la Mama/patología , Replicación Viral , Timidina Quinasa
4.
Cancer Invest ; 30(3): 243-50, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22360364

RESUMEN

Herpes-mediated viral oncolysis alone is not sufficient to completely eradicate tumors. In this study we used a replication conditional, endostatin-expressing herpes simplex virus-1 mutant (HSV-Endo) in a murine lung cancer model. We hypothesized that the anti-angiogenic action of endostatin would improve upon the oncolytic effect of HSV-1. HSV-Endo was evaluated in a pulmonary metastases and orthotopic flank model, where there was significantly less tumor burden and reduced microvessel density compared to a control virus. Endostatin expression appears to improve the anti-tumor effect of HSV-1 in a lung cancer model.


Asunto(s)
Endostatinas/genética , Neoplasias Pulmonares/irrigación sanguínea , Neovascularización Patológica/terapia , Viroterapia Oncolítica/métodos , Simplexvirus/genética , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Células Endoteliales/fisiología , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos BALB C , Transgenes
5.
Bioorg Med Chem Lett ; 22(5): 1958-62, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22318160

RESUMEN

Group II metabotropic glutamate receptors (mGluRs) have been implicated in a variety of neurological and psychiatric disorders in recent studies. As a noninvasive medical imaging technique and a powerful tool in neurological research, positron emission tomography (PET) offers the possibility to visualize and study group II mGluRs in vivo under physiologic and pathologic conditions. We synthesized a PET tracer, (S,S,S)-2-(2-carboxycyclopropyl)-2-(3-[(11)C]methoxyphenethyl) glycine dimethyl ester ([(11)C]CMGDE), as a prodrug for group II mGluRs, and studied its preliminary biological properties in Sprague-Dawley rats to visualize group II mGluRs. The microPET studies demonstrated that [(11)C]CMGDE readily penetrated into the brain and the radiotracer generated from [(11)C]CMGDE had fast reversible binding in the group II mGluRs rich regions including striatum, hippocampus and different cortical areas. Blocking studies with LY341495 showed 20-30% decrease of binding of the radiotracer generated from [(11)C]CMGDE in all brain areas with the highest decrease in the striatum 31.5±3.2%. The results show [(11)C]CMGDE is the first PET tracer that is brain penetrating and can be used to image group II mGluRs in vivo.


Asunto(s)
Encéfalo/diagnóstico por imagen , Tomografía de Emisión de Positrones/métodos , Profármacos/síntesis química , Radiofármacos/síntesis química , Receptores de Glutamato Metabotrópico/análisis , Aminoácidos/farmacología , Animales , Encéfalo/metabolismo , Cuerpo Estriado/diagnóstico por imagen , Cuerpo Estriado/metabolismo , Hipocampo/diagnóstico por imagen , Hipocampo/metabolismo , Masculino , Profármacos/farmacocinética , Unión Proteica/efectos de los fármacos , Radiofármacos/farmacocinética , Ratas , Ratas Sprague-Dawley , Xantenos/farmacología
6.
Sci Rep ; 11(1): 15897, 2021 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-34354107

RESUMEN

Fragile X Syndrome (FXS) is a neurodevelopmental disorder caused by silencing of the Fragile X Mental Retardation (FMR1) gene. The resulting loss of Fragile X Mental Retardation Protein (FMRP) leads to excessive glutamate signaling via metabotropic glutamate subtype 5 receptors (mGluR5) which has been implicated in the pathogenesis of the disorder. In the present study we used the radioligand 3-[18F]fluoro-5-(2-pyridinylethynyl)benzonitrile ([18F]FPEB) in simultaneous PET-MR imaging of males with FXS and age- and gender-matched controls to assess the availability of mGlu5 receptors in relevant brain areas. Patients with FXS showed lower [18F]FPEB binding potential (p < 0.01), reflecting reduced mGluR5 availability, than the healthy controls throughout the brain, with significant group differences in insula, anterior cingulate, parahippocampal, inferior temporal and olfactory cortices, regions associated with deficits in inhibition, memory, and visuospatial processes characteristic of the disorder. The results are among the first to provide in vivo evidence of decreased availability of mGluR5 in the brain in individuals with FXS than in healthy controls. The consistent results across the subjects, despite the tremendous challenges with neuroimaging this population, highlight the robustness of the protocol and support for its use in drug occupancy studies; extending our radiotracer development and application efforts from mice to humans.


Asunto(s)
Síndrome del Cromosoma X Frágil/diagnóstico por imagen , Síndrome del Cromosoma X Frágil/genética , Receptor del Glutamato Metabotropico 5/genética , Adulto , Biomarcadores/metabolismo , Encéfalo/metabolismo , Corteza Cerebral/metabolismo , Radioisótopos de Flúor , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Expresión Génica/genética , Regulación de la Expresión Génica/genética , Humanos , Masculino , Persona de Mediana Edad , Tomografía de Emisión de Positrones/métodos , Receptor del Glutamato Metabotropico 5/metabolismo
7.
J Neurosci ; 29(16): 5193-201, 2009 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-19386915

RESUMEN

The insulin-sensitive isoform of the glucose transporting protein, Glut4, is expressed in fat as well as in skeletal and cardiac muscle and is responsible for the effect of insulin on blood glucose clearance. Recent studies have revealed that Glut4 is also expressed in the brain, although the intracellular compartmentalization and regulation of Glut4 in neurons remains unknown. Using sucrose gradient centrifugation, immunoadsorption and immunofluorescence staining, we have shown that Glut4 in the cerebellum is localized in intracellular vesicles that have the sedimentation coefficient, the buoyant density, and the protein composition similar to the insulin-responsive Glut4-storage vesicles from fat and skeletal muscle cells. In cultured cerebellar neurons, insulin stimulates glucose uptake and causes translocation of Glut4 to the cell surface. Using 18FDG (18fluoro-2-deoxyglucose) positron emission tomography, we found that physical exercise acutely increases glucose uptake in the cerebellum in vivo. Prolonged physical exercise increases expression of the Glut4 protein in the cerebellum. Our results suggest that neurons have a novel type of translocation-competent vesicular compartment which is regulated by insulin and physical exercise similar to Glut4-storage vesicles in peripheral insulin target tissues.


Asunto(s)
Cerebelo/fisiología , Transportador de Glucosa de Tipo 4/fisiología , Insulina/fisiología , Neuronas/fisiología , Vesículas Sinápticas/fisiología , Tejido Adiposo/química , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Animales , Células Cultivadas , Cerebelo/citología , Cerebelo/metabolismo , Ratones , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiología , Neuronas/citología , Neuronas/metabolismo , Transporte de Proteínas/fisiología , Ratas , Ratas Sprague-Dawley , Vesículas Sinápticas/química , Vesículas Sinápticas/metabolismo
8.
Cancer Res ; 67(7): 3295-300, 2007 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-17409438

RESUMEN

Viral oncolysis, the destruction of cancer cells by replicating viruses, is under clinical investigation for cancer therapy. Lytic viral replication in cancer cells both destroys the cells and liberates progeny virion to infect adjacent cancer cells. The safety and efficacy of this approach are dependent on selective and robust viral replication in cancer cells rather than in normal cells. Methods to detect and quantify viral replication in tissues have relied on organ sampling for molecular analyses. Preclinical and clinical studies of viral oncolysis will benefit significantly from development of a noninvasive method to repetitively measure viral replication. We have shown that positron emission tomography (PET) allows for in vivo detection of herpes simplex virus (HSV)-1 replication in tumor cells using 9-(4-[(18)F]-fluoro-3-[hydroxymethyl]butyl)guanine ([(18)F]FHBG) as the substrate for HSV thymidine kinase (HSV-TK). As expected, phosphorylated [(18)F]FHBG is initially trapped within HSV-1-infected tumor cells and is detectable as early as 2 h following virus administration. MicroPET images reveal that [(18)F]FHBG accumulation in HSV-1-infected tumors peaks at 6 h. However, despite progressive accumulation of HSV-1 titers and HSV-TK protein in the tumor as viral oncolysis proceeds, tumor cell degradation resulting from viral oncolysis increases over time, which limits intracellular retention of [(18)F]FHBG. These observations have important consequences with regard to strategies to use [(18)F]FHBG PET for monitoring sites of HSV-TK expression during viral oncolysis.


Asunto(s)
Neoplasias del Colon/terapia , Neoplasias del Colon/virología , Herpesvirus Humano 1/fisiología , Viroterapia Oncolítica/métodos , Animales , Procesos de Crecimiento Celular/fisiología , Chlorocebus aethiops , Neoplasias del Colon/diagnóstico por imagen , Neoplasias del Colon/patología , Radioisótopos de Flúor/farmacocinética , Guanina/análogos & derivados , Guanina/farmacocinética , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Ratones , Tomografía de Emisión de Positrones/métodos , Radiofármacos/farmacocinética , Timidina Quinasa/biosíntesis , Timidina Quinasa/genética , Timidina Quinasa/metabolismo , Replicación Viral
9.
Cancer Gene Ther ; 26(5-6): 145-156, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30420717

RESUMEN

Meningeal metastasis is a fatal complication of breast cancer which affects 8-15% of patients who experience severe neurological complications of cranial nerves, cerebrum, and spinal cord. Survival once diagnosed is less than 4 months. Currently there is no cure. Aggressive multimodal radiation, intra-CSF, or systemic chemotherapy is palliative. Investigation of urgently needed new treatment modalities is hindered by the lack of suitable animal models to effectively study tumor growth kinetics. We present a model of meningeal metastases where tumor growth and associated neurological symptoms have been characterized over 3 weeks by sequential molecular imaging, tumor growth kinetics, and histopathology. Meningeal metastases were induced by stereotaxic injection of human breast cancer cells (MDA-MB-231-Rluc) into the lateral ventricle. Tumor identified by Gd-MRI and Rluc-bioluminescence depict growth in 3 phases, namely lag, exponential, and plateau phase. Invasive tumor growth was highlighted by changes in contrast distribution in the meninges, ventricle and brain compartments over time where moderate contrast uptake in the early growth phase gave rise to a heavy tumor burden in the base of the brain in the latter phases. Tumor growth was accompanied with debilitating neurological symptoms and change in body mass. Tumor was confirmed by ex vivo histology. The reliability of the model to study novel therapeutics was confirmed by oncolytic virus delivered into the lateral ventricle showed potential for treatment. This effective and reliable model resembles human disease progression and is ideally suited to investigate novel treatments.


Asunto(s)
Neoplasias de la Mama/complicaciones , Neoplasias Meníngeas/secundario , Imagen Molecular/métodos , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Meníngeas/patología , Ratones , Ratones Desnudos , Metástasis de la Neoplasia
10.
JAMA ; 299(1): 53-60, 2008 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-18167406

RESUMEN

CONTEXT: Overexpression of epidermal growth factor (EGF) in the liver induces transformation to hepatocellular carcinoma in animal models. Polymorphisms in the EGF gene modulate EGF levels. OBJECTIVE: To assess the relationship among human EGF gene single-nucleotide polymorphism, EGF expression, and risk of hepatocellular carcinoma. DESIGN, SETTING, AND PARTICIPANTS: Molecular mechanisms linking the 61*G allele polymorphism to EGF expression were examined in human hepatocellular carcinoma cell lines and human liver tissue. A case-control study involving 207 patients with cirrhosis was conducted at the Massachusetts General Hospital (1999-2006) and a validation case-control study involving 121 patients with cirrhosis was conducted at Hôpital Paul Brousse (1993-2006). Restriction fragment-length polymorphism was used to determine the EGF gene polymorphism genotype. Logistic regression analysis was used to assess the association between the EGF polymorphism and hepatocellular carcinoma risk. MAIN OUTCOME MEASURES: Mechanisms by which the EGF gene polymorphism modulates EGF levels and associations among EGF gene polymorphism, EGF levels, and hepatocellular carcinoma. RESULTS: Transcripts from the EGF 61*G allele exhibited more than a 2-fold longer half-life than those from the 61*A allele, and EGF secretion was 2.3-fold higher in G/G hepatocellular carcinoma cell lines than A/A cell lines. Serum EGF levels were 1.8-fold higher in G/G patients than A/A patients, and liver EGF levels were 2.4-fold higher in G/G patients than A/A patients. Among the 207 patients with cirrhosis in the Massachusetts study population, 59 also had hepatocellular carcinoma. Analysis of the distribution of allelic frequencies revealed that there was a 4-fold odds of hepatocellular carcinoma in G/G patients compared with A/A patients in the Massachusetts study population (odds ratio, 4.0; 95% confidence interval [CI], 1.6-9.6; P = .002). Logistic regression analysis demonstrated that the number of copies of G was significantly associated with hepatocellular carcinoma after adjusting for age, sex, race, etiology, and severity of cirrhosis (G/G or A/G vs A/A; hazard ratio, 3.49; 95% CI, 1.29-9.44; P = .01). The significant association was validated in the French patients with alcoholic cirrhosis and hepatocellular carcinoma. CONCLUSION: The EGF gene polymorphism genotype is associated with risk for development of hepatocellular carcinoma in liver cirrhosis through modulation of EGF levels.


Asunto(s)
Carcinoma Hepatocelular/genética , Factor de Crecimiento Epidérmico/genética , Cirrosis Hepática/complicaciones , Neoplasias Hepáticas/genética , Polimorfismo de Nucleótido Simple , Carcinoma Hepatocelular/etiología , Carcinoma Hepatocelular/metabolismo , Estudios de Casos y Controles , Línea Celular Tumoral , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Factor de Crecimiento Epidérmico/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Genotipo , Humanos , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Neoplasias Hepáticas/etiología , Neoplasias Hepáticas/metabolismo , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Factores de Riesgo
12.
Cancer Res ; 65(9): 3823-7, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15867380

RESUMEN

Dog and rat animal models have been developed for repeated intravascular administrations to the liver. However, mice have generally been considered too small to use for these models. This study describes the development of mouse models that permit the establishment of liver metastases that can be subsequently treated by repeated injections into the portal venous system. A mini-laparotomy is done to mobilize the spleen and transpose it to a s.c. pocket with its vascular pedicle intact. A suspension of single tumor cells is then inoculated into the portal vein to establish diffuse liver metastases. These tumors may be treated by simple percutaneous injections directly into the s.c. whole spleen reservoir. The ease of injection into the s.c. spleen permits repeated injections into the portal venous system. The usefulness of this model was shown in experiments revealing that multiple portal venous administrations of a replication-conditional, oncolytic herpes simplex virus mutant are more effective than a single portal venous administration. In a modification of this model, the spleen is first split into two, leaving intact the vascular pedicle for each half of the spleen. Tumor cells are inoculated into one hemi-spleen, which is then resected 10 minutes later. The other hemi-spleen is transposed to the s.c. position, thereby permitting subsequent repetitive portal venous injections via percutaneous injections into the s.c. hemi-spleen. These mouse models are useful for a wide range of studies.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Neoplasias Hepáticas Experimentales/secundario , Neoplasias Hepáticas Experimentales/virología , Hígado/irrigación sanguínea , Simplexvirus/fisiología , Bazo/irrigación sanguínea , Bazo/cirugía , Animales , Chlorocebus aethiops , Modelos Animales de Enfermedad , Inyecciones Intravenosas , Neoplasias Hepáticas Experimentales/irrigación sanguínea , Neoplasias Hepáticas Experimentales/tratamiento farmacológico , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias/métodos , Vena Porta , Bazo/patología , Células Vero
13.
Neuropharmacology ; 108: 462-73, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26581500

RESUMEN

G-protein coupled dopamine and metabotropic glutamate receptors (mGlu) can modulate neurotransmission during Parkinson's disease (PD)-like neurodegeneration. PET imaging studies in a unilateral dopamine denervation model (6-OHDA) showed a significant inverse correlation of presynaptic mGlu4 and postsynaptic mGlu5 expression in the striatum and rapidly declining mGlu4 and enhanced mGlu5 expression in the hippocampus during progressive degeneration over time. Immunohistochemical studies verified the decreased mGlu4 expression in the hippocampus on the lesion side but did not show difference in mGlu5 expression between lesion and control side. Pharmacological MRI studies showed enhanced hemodynamic response in several brain areas on the lesion side compared to the control side after challenge with mGlu4 positive allosteric modulator or mGlu5 negative allosteric modulator. However, mGlu4 response was biphasic having short enhancement followed by negative response on both sides of brain. Studies in mGlu4 expressing cells demonstrated that glutamate induces cooperative increase in binding of mGlu4 ligands - especially at high glutamate levels consistent with in vivo concentration. This suggests that mGlu allosteric modulators as drug candidates will be highly sensitive to changes in glutamate concentration and hence metabolic state. These experiments demonstrate the importance of the longitudinal imaging studies to investigate temporal changes in receptor functions to obtain individual response for experimental drugs.


Asunto(s)
Enfermedades Neurodegenerativas/diagnóstico por imagen , Enfermedades Neurodegenerativas/metabolismo , Trastornos Parkinsonianos/diagnóstico por imagen , Trastornos Parkinsonianos/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Animales , Células CHO , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Ácido Glutámico/farmacología , Masculino , Unión Proteica/fisiología , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Glutamato Metabotrópico/metabolismo
14.
Cancer Biol Ther ; 4(5): 524-31, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15917655

RESUMEN

The use of viruses to destroy tumors, also known as viral oncolysis, dates back to the early 1900's. Although the mechanism of cancer cell lysis was unknown in the early years of development, advances in tumor biology, molecular biology, and virology have been critical for numerous advances that have brought the field to where it is today. Oncolytic viruses have been developed based on innate and engineered properties to preferentially target tumor cells. Engineered properties include alterations in endogenous gene expression and introduction of foreign genes. Methods to non-invasively monitor sites of viral replication is required for preclinical and clinical studies. Positron emission tomography (PET) can be used for this purpose. This review focuses on commonly used oncolytic viruses, their selection for oncolytic therapy, the design of HSV-1 viral mutants, and monitoring their replication by PET.


Asunto(s)
Terapia Genética , Herpesvirus Humano 1/fisiología , Virus Oncolíticos/fisiología , Animales , Ingeniería Genética , Herpesvirus Humano 1/genética , Humanos , Mutación , Neoplasias/terapia , Neoplasias/virología , Virus Oncolíticos/genética , Tomografía de Emisión de Positrones
15.
Neuropharmacology ; 97: 142-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26025660

RESUMEN

The interest in the role of metabotropic glutamate receptor 4 (mGlu4) in CNS related disorders has increased the need for methods to investigate the binding of allosteric drug candidates. Our aim is to present the first fully characterized in vitro binding assay of mGlu4 positive allosteric modulators (PAMs). Results suggest that mGlu4 PAMs have characteristic co-operative binding with orthosteric glutamate, which offers a notable insight to the further development of mGlu4 targeted therapies.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Receptores de Glutamato Metabotrópico/metabolismo , Regulación Alostérica , Animales , Unión Competitiva , Células CHO , Cricetulus , Relación Dosis-Respuesta a Droga , Fármacos actuantes sobre Aminoácidos Excitadores/síntesis química , Fármacos actuantes sobre Aminoácidos Excitadores/química , Ácido Glutámico/metabolismo
16.
Cancer Res ; 74(15): 4111-21, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24876106

RESUMEN

Viral oncolysis, the destruction of cancer cells by replicating virus, is an experimental cancer therapy that continues to be explored. The treatment paradigm for this therapy involves successive waves of lytic replication in cancer cells. At present, monitoring viral titer at sites of replication requires biopsy. However, repeat serial biopsies are not practically feasible for temporal monitoring of viral replication and tumor response in patients. Molecular imaging provides a noninvasive method to identify intracellular viral gene expression in real time. We imaged viral oncolysis and tumor response to oncolysis sequentially with bioluminescence and positron emission tomography (PET), revealing the kinetics of both processes in tumor xenografts. We demonstrate that virus replication cycles can be identified as successive waves of reporter expression that occur ∼2 days after the initial viral tumor infection peak. These waves correspond to virions that are released following a replication cycle. The viral and cellular kinetics were imaged with Fluc and Rluc bioluminescence reporters plus two 18F-labeled PET reporters FHBG [9-(4-18F-fluoro-3-[hydroxymethyl] butyl) guanine] and FLT (18F-3'-deoxy-3-'fluorothymidine), respectively. Correlative immunohistochemistry on tumor xenograft sections confirmed in vivo results. Our findings show how PET can be used to identify virus replication cycles and for real-time measurements of intratumoral replicating virus levels. This noninvasive imaging approach has potential utility for monitoring viral oncolysis therapy in patients.


Asunto(s)
Mediciones Luminiscentes/métodos , Imagen Molecular/métodos , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Tomografía de Emisión de Positrones/métodos , Animales , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Neoplasias de la Mama/virología , Línea Celular Tumoral , Chlorocebus aethiops , Neoplasias del Colon/diagnóstico por imagen , Neoplasias del Colon/patología , Neoplasias del Colon/terapia , Neoplasias del Colon/virología , Femenino , Células HT29 , Humanos , Melanoma Amelanótico/diagnóstico por imagen , Melanoma Amelanótico/patología , Melanoma Amelanótico/terapia , Melanoma Amelanótico/virología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células Vero
17.
J Nucl Med ; 54(2): 252-8, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23376854

RESUMEN

UNLABELLED: Over the last several years, epidemiologic data have suggested that the antidiabetes drug metformin (MET), an adenosine monophosphate-activated protein kinase (AMPK) activator, improves progression-free survival of patients with multiple cancers; more than 30 clinical trials are under way to confirm this finding. We postulated that the role of AMPK as a central cellular energy sensor would result in opposite effects on glucose uptake and proliferation, suggesting different roles for (18)F-FDG and 3'-deoxy-3'-(18)F-fluorothymidine ((18)F-FLT) in assessing its effectiveness as an antineoplastic agent. METHODS: Colon cancer cell lines HT29 (human) and MC26 (murine) were treated for 24 or 72 h with a range of MET concentrations (0-10 mM). Western blotting was used to study the activation of AMPK after MET treatment. Glucose uptake and cell proliferation were measured by cell retention studies with either (18)F-FDG or (18)F-FLT. EdU (ethynyl deoxyuridine, a thymidine analog) and annexin-propidium iodide flow cytometry was performed to determine cell cycle S-phase and apoptotic changes. In vivo (18)F-FDG and (18)F-FLT PET images were acquired before and 24 h after MET treatment of HT29 tumor-bearing mice. RESULTS: After 24 h of MET incubation, phosphorylated AMPK levels increased severalfold in both cell lines, whereas total AMPK levels remained unchanged. In cell retention studies, (18)F-FDG uptake increased but (18)F-FLT retention decreased significantly in both cell lines. The numbers of HT29 and MC26 cells in the S phase decreased 36% and 33%, respectively, after MET therapy. Apoptosis increased 10.5-fold and 5.8-fold in HT29 and MC26 cells, respectively, after 72 h of incubation with MET. PET imaging revealed increased (18)F-FDG uptake (mean ± SEM standardized uptake values were 0.71 ± 0.03 before and 1.29 ± 0.11 after MET therapy) (P < 0.05) and decreased (18)F-FLT uptake (mean ± SEM standardized uptake values were 1.18 ± 0.05 before and 0.89 ± 0.01 after MET therapy) (P < 0.05) in HT29 tumor-bearing mice. CONCLUSION: MET, through activation of the AMPK pathway, produces a dose-dependent increase in tumor glucose uptake while decreasing cell proliferation in human and murine colon cancer cells. Thus, changes in (18)F-FDG uptake after MET treatment may be misleading. (18)F-FLT imaging is a promising alternative that correlates with the tumor response.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Fluorodesoxiglucosa F18/farmacología , Metformina/farmacología , Neoplasias/tratamiento farmacológico , Tomografía de Emisión de Positrones/métodos , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Apoptosis , Ciclo Celular , Línea Celular Tumoral , Supervivencia sin Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Ratones , Ratones Desnudos , Modelos Biológicos , Factores de Tiempo
18.
PLoS One ; 8(3): e58290, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23505478

RESUMEN

Here we introduce diffusion molecular retention (DMR) tumor targeting, a technique that employs PEG-fluorochrome shielded probes that, after a peritumoral (PT) injection, undergo slow vascular uptake and extensive interstitial diffusion, with tumor retention only through integrin molecular recognition. To demonstrate DMR, RGD (integrin binding) and RAD (control) probes were synthesized bearing DOTA (for (111) In(3+)), a NIR fluorochrome, and 5 kDa PEG that endows probes with a protein-like volume of 25 kDa and decreases non-specific interactions. With a GFP-BT-20 breast carcinoma model, tumor targeting by the DMR or i.v. methods was assessed by surface fluorescence, biodistribution of [(111)In] RGD and [(111)In] RAD probes, and whole animal SPECT. After a PT injection, both probes rapidly diffused through the normal and tumor interstitium, with retention of the RGD probe due to integrin interactions. With PT injection and the [(111)In] RGD probe, SPECT indicated a highly tumor specific uptake at 24 h post injection, with 352%ID/g tumor obtained by DMR (vs 4.14%ID/g by i.v.). The high efficiency molecular targeting of DMR employed low probe doses (e.g. 25 ng as RGD peptide), which minimizes toxicity risks and facilitates clinical translation. DMR applications include the delivery of fluorochromes for intraoperative tumor margin delineation, the delivery of radioisotopes (e.g. toxic, short range alpha emitters) for radiotherapy, or the delivery of photosensitizers to tumors accessible to light.


Asunto(s)
Colorantes Fluorescentes/metabolismo , Neoplasias/metabolismo , Animales , Línea Celular Tumoral , Difusión , Femenino , Colorantes Fluorescentes/administración & dosificación , Colorantes Fluorescentes/química , Genes Reporteros , Humanos , Inyecciones , Integrinas/metabolismo , Ratones , Imagen Molecular , Imagen Multimodal , Neoplasias/diagnóstico , Polietilenglicoles/química , Tomografía de Emisión de Positrones , Radiofármacos/administración & dosificación , Radiofármacos/química , Radiofármacos/metabolismo , Distribución Tisular , Tomografía Computarizada por Rayos X , Trasplante Heterólogo
19.
Cancer Res ; 70(3): 948-56, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20103648

RESUMEN

Metastasis relies on angiogenesis for tumor expansion. Tumor angiogenesis is restrained by a variety of endogenous inhibitors, including thrombospondin 1 (TSP1). The principal antiangiogenic activity of TSP1 resides in a domain containing three TSP1 repeats (3TSR), and TSP1 cleavage is regulated, in part, by the metalloproteinase ADAMTS1. In this study, we examined the role of TSP1 and ADAMTS1 in controlling metastatic disease in the liver and lung. TSP1 overexpression inhibited metastatic growth of colon or renal carcinoma cells in liver but not lung. Metastatic melanoma in liver grew more rapidly in Tsp1-null mice compared with controls, whereas in lung grew similarly in Tsp1-null mice or controls. Recombinant TSP1 was cleaved more efficiently in lysates from liver than lung. ADAMTS1 inhibition by neutralizing antibody, small interfering RNA, or genetic deletion abrogated cleavage activity. To confirm that lack of cleavage of TSP1 ablated its antiangiogenic function in the lung, we generated colon cancer cells stably secreting only the 3TSR domain and found that they inhibited formation of both liver and lung metastases. Collectively, our results indicate that the antiangiogenic activity of TSP1 is differentially regulated by ADAMTS1 in the liver and lung, emphasizing the concept that regulation of angiogenesis is varied in different tissue environments.


Asunto(s)
Proteínas ADAM/metabolismo , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/secundario , Neoplasias Experimentales/patología , Trombospondina 1/metabolismo , Proteínas ADAM/genética , Proteína ADAMTS1 , Animales , Sitios de Unión , Western Blotting , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trombospondina 1/genética , Transfección
20.
Am J Physiol Heart Circ Physiol ; 295(6): H2495-502, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18978196

RESUMEN

Insulin resistance is an increasingly prevalent condition in humans that frequently clusters with disorders characterized by left ventricular (LV) pressure overload, such as systemic hypertension. To investigate the impact of insulin resistance on LV remodeling and functional response to pressure overload, C57BL6 male mice were fed a high-fat (HFD) or a standard diet (SD) for 9 days and then underwent transverse aortic constriction (TAC). LV size and function were assessed in SD- and HFD-fed mice using serial echocardiography before and 7, 21, and 28 days after TAC. Serial echocardiography was also performed on nonoperated SD- and HFD-fed mice over a period of 6 wk. LV perfusion was assessed before and 7 and 28 days after TAC. Nine days of HFD induced systemic and myocardial insulin resistance (assessed by myocardial 18F-fluorodeoxyglucose uptake), and myocardial perfusion response to acetylcholine was impaired. High-fat feeding for 28 days did not change LV size and function in nonbanded mice; however, TAC induced greater hypertrophy, more marked LV systolic and diastolic dysfunction, and decreased survival in HFD-fed compared with SD-fed mice. Compared with SD-fed mice, myocardial perfusion reserve was decreased 7 days after TAC, and capillary density was decreased 28 days after TAC in HFD-fed mice. A short duration of HFD induces insulin resistance in mice. These metabolic changes are accompanied by increased LV remodeling and dysfunction after TAC, highlighting the impact of insulin resistance in the development of pressure-overload-induced heart failure.


Asunto(s)
Circulación Coronaria , Grasas de la Dieta/efectos adversos , Insuficiencia Cardíaca/etiología , Hipertensión/complicaciones , Resistencia a la Insulina , Disfunción Ventricular Izquierda/etiología , Remodelación Ventricular , Acetilcolina/farmacología , Animales , Apoptosis , Glucemia/metabolismo , Capilares/patología , Capilares/fisiopatología , Circulación Coronaria/efectos de los fármacos , Modelos Animales de Enfermedad , Dobutamina/farmacología , Fibrosis , Insuficiencia Cardíaca/diagnóstico por imagen , Insuficiencia Cardíaca/fisiopatología , Hemodinámica , Hipertensión/diagnóstico por imagen , Hipertensión/fisiopatología , Insulina/sangre , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Endogámicos C57BL , Imagen de Perfusión Miocárdica , Miocardio/metabolismo , Miocardio/patología , Tomografía de Emisión de Positrones , Factores de Riesgo , Factores de Tiempo , Ultrasonografía , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Disfunción Ventricular Izquierda/diagnóstico por imagen , Disfunción Ventricular Izquierda/fisiopatología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda