Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Clin Cancer Res ; 30(6): 1152-1159, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38236581

RESUMEN

PURPOSE: HOXB13 is an androgen receptor (AR) coregulator specifically expressed in cells of prostatic lineage. We sought to associate circulating tumor cell (CTC) HOXB13 expression with outcomes in men with mCRPC treated with abiraterone or enzalutamide. EXPERIMENTAL DESIGN: We conducted a retrospective analysis of the multicenter prospective PROPHECY trial of mCRPC men (NCT02269982, n = 118) treated with abiraterone/enzalutamide. CTC detection and HOXB13 complementary DNA (cDNA) expression was measured using a modified Adnatest, grouping patients into 3 categories: CTC 0 (undetectable); CTC+ HOXB13 CTC low (<4 copies); or CTC+ HOXB13 CTC high. The HOXB13 threshold was determined by maximally selected rank statistics for prognostic associations with overall survival (OS) and progression-free survival (PFS). RESULTS: We included 102 men with sufficient CTC HOXB13 cDNA, identifying 25%, 31%, and 44% of patients who were CTC 0, CTC+ HOXB13 low, and CTC+ HOXB13 high, respectively. Median OS were 25.7, 27.8, and 12.1 months whereas the median PFS were 9.0, 7.7, and 3.8 months, respectively. In subgroup analysis among men with CellSearch CTCs ≥5 copies/mL and adjusting for prior abi/enza treatment and Halabi clinical risk score, the multivariate HR for HOXB13 CTC detection was 2.39 (95% CI, 1.06-5.40) for OS and 2.78 (95% CI, 1.38-5.59) for PFS, respectively. Low HOXB13 CTC detection was associated with lower CTC PSA, PSMA, AR-FL, and AR-V7 detection, and more liver/lung metastases (41% vs. 25%). CONCLUSIONS: Higher CTC HOXB13 expression is associated with AR-dependent biomarkers in CTCs and is adversely prognostic in the context of potent AR inhibition in men with mCRPC.


Asunto(s)
Androstenos , Benzamidas , Células Neoplásicas Circulantes , Feniltiohidantoína , Neoplasias de la Próstata Resistentes a la Castración , Masculino , Humanos , Células Neoplásicas Circulantes/patología , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , ARN , Estudios Prospectivos , Estudios Retrospectivos , ADN Complementario/uso terapéutico , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Nitrilos/uso terapéutico , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/uso terapéutico , Proteínas de Homeodominio/genética
2.
Biomed Pharmacother ; 168: 115792, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37924789

RESUMEN

Ulipristal acetate (UPA) is a selective progesterone receptor modulator and is used for the treatment of uterine leiomyoma (a benign tumor). Uterine sarcoma which is highly malignant cancer with a poor prognosis is clinically resembled with uterine leiomyoma. There has been no experimental research on the effect of UPA on uterine sarcoma. In this study, we examined the efficacy of UPA in uterine sarcoma with in vitro and in vivo animal models. Cytotoxicity of UPA was determined in uterine sarcoma cell lines (MES-SA, SK-UT-1, and SK-LMS-1). Apoptotic genes and signaling pathways affected by UPA were analyzed by complementary DNA (cDNA) microarray of uterine sarcoma cell lines and western blot, respectively. An in vivo efficacy of UPA was examined with uterine sarcoma cell line- and patient-derived xenograft (PDX) mice models. UPA inhibited cell growth in uterine sarcoma cell lines and primary culture cells from a PDX mouse (PDX-C). cDNA microarray analysis revealed that CCL2 was highly down-regulated by UPA. Phosphorylation and the total expression of STAT3 were inhibited by UPA. UPA also inhibited CCL2 and STAT3 in PDX-C. The inhibitory effect of UPA had not changed in the overexpression of PR and treatment of progesterone. In vivo efficacy studies with cell line-derived xenografts and a PDX model with leiomyosarcoma, a typical uterine sarcoma, demonstrated that UPA significantly decreased tumor growth. UPA had significant anti-tumor effects in uterine sarcoma through the inhibition of STAT3/CCL2 signaling pathway and might be a potential therapeutic agent to treat this disease.


Asunto(s)
Leiomioma , Sarcoma , Neoplasias Uterinas , Femenino , Humanos , Animales , Ratones , Receptores de Progesterona/metabolismo , ADN Complementario/farmacología , ADN Complementario/uso terapéutico , Neoplasias Uterinas/patología , Leiomioma/patología , Transducción de Señal , Muerte Celular , Sarcoma/tratamiento farmacológico , Quimiocina CCL2/metabolismo , Factor de Transcripción STAT3/metabolismo
3.
Front Endocrinol (Lausanne) ; 13: 950345, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36120434

RESUMEN

Due to the lack of symptoms and detection biomarkers at the early stage, most patients with ovarian cancer (OC) are diagnosed at an advanced stage and often face chemoresistance and relapse. Hence, defining detection biomarkers and mechanisms of chemoresistance is imperative. A previous report of a cDNA microarray analysis shows a potential association of carnitine O-octanoyltransferase (CROT) with taxane resistance but the biological function of CROT in OC remains unknown. The current study explored the function and regulatory mechanism of CROT on cellular behavior and paclitaxel (PTX)-resistance in OC. We found that CROT was downregulated in OC tissues and PTX-resistant cells. Furthermore, CROT expression was negatively correlated with the prognosis of OC patients. Overexpression of CROT inhibited the OC cell proliferation, migration, invasion, and colony formation, arrested the cell cycle at the G2/M phase, and promoted cell apoptosis. In addition, miR-33a-5p bound directly to the 3'UTR of CROT to negatively regulate the expression of CROT and promoted OC cell growth. Finally, overexpression of CROT decreased the phosphorylation of Smad2, whereas knockdown of CROT increased the nuclear translocation of Smad2 and Smad4, two transducer proteins of TGF-ß signaling, indicating that CROT is a tumor suppressor which mediates OC cell behaviors through the TGF-ß signaling pathway. Thus, targeting the miR-33a-5p/CROT axis may have clinical potential for the treatment of patients with OC.


Asunto(s)
MicroARNs , Neoplasias Ováricas , Regiones no Traducidas 3' , Carnitina , Línea Celular Tumoral , ADN Complementario/uso terapéutico , Resistencia a Antineoplásicos/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Recurrencia Local de Neoplasia/genética , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Transducción de Señal , Taxoides/uso terapéutico , Factor de Crecimiento Transformador beta
4.
Turk J Gastroenterol ; 33(8): 696-703, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35943151

RESUMEN

BACKGROUND: First-line treatments for metastatic pancreatic cancer are chemotherapy regimens consisting of 5-fluorouracil or gemcitabine; however, there are no biomarkers to help determine which patients might benefit from which treatment regimens. We aimed to show that microRNAs let-7c and 7d can be used as independent predictive biomarkers for metastatic pancreatic cancer. METHODS: A total of 55 patients who had first-line chemotherapy with FOLFIRINOX or gemcitabine+capecitabine were included. Patients were divided into groups based on let-7c and let-7d levels and chemotherapy treatment as let-7c-7d high FOLFIRINOX, let7c-7d high gemcitabine+capecitabine, let-7c-7d low FOLFIRINOX, and let-7c-7d low gemcitabine+capecitabine. Blood samples were taken from patients before chemotherapy for microRNA let-7c and 7d analysis. MicroRNA isolation was performed using a miRNeasy Serum/Plasma Kit and identified using spectrophotometric measurements. After isolation, microRNA was converted to cDNA using a microRNA cDNA Synthesis Kit with poly (A) polymerase tailing. The expression of microRNA was examined using quantitative real-time polymerase chain reaction. RESULTS: The overall survival of patients who received FOLFIRINOX treatment with a high let-7c-7d level was statistically significantly longer than those who received gemcitabine+capecitabine with a high let-7c-7d level. In addition, patients with low let-7c expression receiving FOLFIRINOX progressed significantly 2.104 times earlier than patients with high let-7c expression receiving FOLFIRINOX. CONCLUSION: The serum MicroRNA let-7c level was found to be an independent predictive biomarker in the FOLFIRINOX treatment group.


Asunto(s)
MicroARNs , Neoplasias Pancreáticas , Humanos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores , Capecitabina/uso terapéutico , ADN Complementario/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , ARN Mensajero/metabolismo
5.
Hum Gene Ther Methods ; 27(5): 174-186, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27477497

RESUMEN

Duchenne muscular dystrophy (DMD) is a severe, genetic muscle disease caused by the absence of the sarcolemmal protein dystrophin. Gene replacement therapy is considered a potential strategy for the treatment of DMD, aiming to restore the missing protein. Although the elements of the dystrophin molecule have been identified and studies in transgenic mdx mice have explored the importance of a number of these structural domains, the resulting modified dystrophin protein products that have been developed so far are only partially characterized in relation to their structure and function in vivo. To optimize a dystrophin cDNA construct for therapeutic application we designed and produced four human minidystrophins within the packaging capacity of lentiviral vectors. Two novel minidystrophins retained the centrally located neuronal nitric oxide synthase (nNOS)-anchoring domain in order to achieve sarcolemmal nNOS restoration, which is lost in most internally deleted dystrophin constructs. Functionality of the resulting truncated dystrophin proteins was investigated in muscle of adult dystrophin-deficient mdx mice followed by a battery of detailed immunohistochemical and morphometric tests. This initial assessment aimed to determine the overall suitability of various constructs for cloning into lentiviral vectors for ex vivo gene delivery to stem cells for future preclinical studies.


Asunto(s)
Distrofina/genética , Terapia Genética , Distrofia Muscular de Duchenne/terapia , Óxido Nítrico Sintasa de Tipo I/genética , Animales , ADN Complementario/genética , ADN Complementario/uso terapéutico , Distrofina/uso terapéutico , Expresión Génica , Vectores Genéticos/uso terapéutico , Humanos , Ratones , Ratones Endogámicos mdx , Ratones Transgénicos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/genética , Óxido Nítrico Sintasa de Tipo I/biosíntesis
6.
Nanomedicine ; 12(3): 811-821, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26518603

RESUMEN

Our goals were to develop and establish nanoparticle (NP)-facilitated inhalational gene delivery, and to validate its biomedical application by testing the hypothesis that targeted upregulation of pulmonary erythropoietin receptor (EpoR) expression protects against lung injury. Poly-lactic-co-glycolic acid (PLGA) NPs encapsulating various tracers were characterized and nebulizated into rat lungs. Widespread NP uptake and distribution within alveolar cells were visualized by magnetic resonance imaging, and fluorescent and electron microscopy. Inhalation of nebulized NPs bearing EpoR cDNA upregulated pulmonary EpoR expression and downstream signal transduction (ERK1/2 and STAT5 phosphorylation) in rats for up to 21 days, and attenuated hyperoxia-induced damage in lung tissue based on apoptosis, oxidative damage of DNA, protein and lipid, tissue edema, and alveolar morphology compared to vector-treated control animals. These results establish the feasibility and therapeutic efficacy of NP-facilitated cDNA delivery to the lung, and demonstrate that targeted pulmonary EpoR upregulation mitigates acute oxidative lung damage. FROM THE CLINICAL EDITOR: Acute lung injury often results in significant morbidity and mortality, and current therapeutic modalities have proven to be ineffective. In this article, the authors developed nanocarrier based gene therapy in an attempt to upregulate the expression of pulmonary erythropoietin receptor in an animal model. Inhalation delivery resulted in reduction of lung damage.


Asunto(s)
ADN Complementario/uso terapéutico , Hiperoxia/terapia , Ácido Láctico/química , Lesión Pulmonar/terapia , Pulmón/patología , Nanopartículas/química , Ácido Poliglicólico/química , Receptores de Eritropoyetina/genética , Administración por Inhalación , Animales , Línea Celular , ADN Complementario/administración & dosificación , ADN Complementario/genética , Técnicas de Transferencia de Gen , Humanos , Hiperoxia/genética , Hiperoxia/patología , Pulmón/metabolismo , Lesión Pulmonar/genética , Lesión Pulmonar/patología , Nanopartículas/ultraestructura , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba
7.
Neurosci Lett ; 590: 74-9, 2015 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-25641132

RESUMEN

The formation of glial scar restricts axon regeneration after spinal cord injury (SCI) in adult mammalian. Chondroitin sulfate proteoglycans (CSPGs) are mostly secreted by reactive astrocytes, which form dense scar tissues after SCI. Chondroitinase ABC (ChABC), which can digest CSPGs, is a promising therapeutic strategy for SCI. However, to date ChABC has exhibited only limited success in the treatment of chronic SCI. The intermediate filament protein vimentin underpins the cytoskeleton of reactive astrocytes. We targeted glial scar in injured spinal cord by sustained infusion of ChABC and antisense vimentin cDNA. Using anterograde tracing, BBB scoring and hind limb placing response, we found that this combined treatment promoted axon regeneration and functional recovery after SCI in rats. Our results indicate that axon regeneration may be promoted by modified physical and biochemical characteristics of intra- and extracellular architecture in glial scar tissues. Theses findings could potentially help us to understand better the composition of glial scar in central nervous system injury.


Asunto(s)
Axones/efectos de los fármacos , Condroitina ABC Liasa/farmacología , ADN sin Sentido/farmacología , ADN Complementario/farmacología , Traumatismos de la Médula Espinal/tratamiento farmacológico , Vimentina/genética , Animales , Axones/fisiología , Condroitina ABC Liasa/uso terapéutico , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , ADN sin Sentido/uso terapéutico , ADN Complementario/uso terapéutico , Quimioterapia Combinada , Femenino , Actividad Motora/efectos de los fármacos , Tractos Piramidales/efectos de los fármacos , Tractos Piramidales/fisiopatología , Tractos Piramidales/ultraestructura , Ratas Sprague-Dawley , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología
8.
N Engl J Med ; 371(15): 1407-17, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25295500

RESUMEN

BACKGROUND: In previous clinical trials involving children with X-linked severe combined immunodeficiency (SCID-X1), a Moloney murine leukemia virus-based γ-retrovirus vector expressing interleukin-2 receptor γ-chain (γc) complementary DNA successfully restored immunity in most patients but resulted in vector-induced leukemia through enhancer-mediated mutagenesis in 25% of patients. We assessed the efficacy and safety of a self-inactivating retrovirus for the treatment of SCID-X1. METHODS: We enrolled nine boys with SCID-X1 in parallel trials in Europe and the United States to evaluate treatment with a self-inactivating (SIN) γ-retrovirus vector containing deletions in viral enhancer sequences expressing γc (SIN-γc). RESULTS: All patients received bone marrow-derived CD34+ cells transduced with the SIN-γc vector, without preparative conditioning. After 12.1 to 38.7 months of follow-up, eight of the nine children were still alive. One patient died from an overwhelming adenoviral infection before reconstitution with genetically modified T cells. Of the remaining eight patients, seven had recovery of peripheral-blood T cells that were functional and led to resolution of infections. The patients remained healthy thereafter. The kinetics of CD3+ T-cell recovery was not significantly different from that observed in previous trials. Assessment of insertion sites in peripheral blood from patients in the current trial as compared with those in previous trials revealed significantly less clustering of insertion sites within LMO2, MECOM, and other lymphoid proto-oncogenes in our patients. CONCLUSIONS: This modified γ-retrovirus vector was found to retain efficacy in the treatment of SCID-X1. The long-term effect of this therapy on leukemogenesis remains unknown. (Funded by the National Institutes of Health and others; ClinicalTrials.gov numbers, NCT01410019, NCT01175239, and NCT01129544.).


Asunto(s)
Gammaretrovirus/genética , Terapia Genética , Vectores Genéticos , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia , Animales , Antígenos CD34 , ADN Complementario/uso terapéutico , Expresión Génica , Silenciador del Gen , Terapia Genética/efectos adversos , Humanos , Lactante , Subunidad gamma Común de Receptores de Interleucina/genética , Masculino , Ratones , Mutación , Linfocitos T/inmunología , Transducción Genética , Transgenes/fisiología , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/inmunología
9.
PLoS One ; 9(8): e104957, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25126862

RESUMEN

Viral vectors have been used for hemophilia A gene therapy. However, due to its large size, full-length Factor VIII (FVIII) cDNA has not been successfully delivered using conventional viral vectors. Moreover, viral vectors may pose safety risks, e.g., adverse immunological reactions or virus-mediated cytotoxicity. Here, we took advantages of the non-viral vector gene delivery system based on piggyBac DNA transposon to transfer the full-length FVIII cDNA, for the purpose of treating hemophilia A. We tested the efficiency of this new vector system in human 293T cells and iPS cells, and confirmed the expression of the full-length FVIII in culture media using activity-sensitive coagulation assays. Hydrodynamic injection of the piggyBac vectors into hemophilia A mice temporally treated with an immunosuppressant resulted in stable production of circulating FVIII for over 300 days without development of anti-FVIII antibodies. Furthermore, tail-clip assay revealed significant improvement of blood coagulation time in the treated mice. piggyBac transposon vectors can facilitate the long-term expression of therapeutic transgenes in vitro and in vivo. This novel gene transfer strategy should provide safe and efficient delivery of FVIII.


Asunto(s)
ADN Complementario/uso terapéutico , Factor VIII/genética , Vectores Genéticos/uso terapéutico , Hemofilia A/terapia , Animales , Elementos Transponibles de ADN , ADN Complementario/administración & dosificación , ADN Complementario/genética , Modelos Animales de Enfermedad , Factor VIII/análisis , Expresión Génica , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Células HEK293 , Hemofilia A/sangre , Hemofilia A/genética , Humanos , Ratones
10.
Am J Vet Res ; 72(12): 1631-8, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22126691

RESUMEN

OBJECTIVE: To evaluate the safety and efficacy of a vaccine containing plasmid DNA with an insert encoding human tyrosinase (ie, huTyr vaccine) as adjunctive treatment for oral malignant melanoma (MM) in dogs. ANIMALS: 111 dogs (58 prospectively enrolled in a multicenter clinical trial and 53 historical controls) with stage II or III oral MM (modified World Health Organization staging scale, I to IV) in which locoregional disease control was achieved. PROCEDURES: 58 dogs received an initial series of 4 injections of huTyr vaccine (102 µg of DNA/injection) administered transdermally by use of a needle-free IM vaccination device. Dogs were monitored for adverse reactions. Surviving dogs received booster injections at 6-month intervals thereafter. Survival time for vaccinates was compared with that of historical control dogs via Kaplan-Meier survival analysis for the outcome of death. RESULTS: Kaplan-Meier analysis of survival time until death attributable to MM was determined to be significantly improved for dogs that received the huTyr vaccine, compared with that of historical controls. However, median survival time could not be determined for vaccinates because < 50% died of MM before the end of the observation period. No systemic reactions requiring veterinary intervention were associated with vaccination. Local reactions were primarily limited to acute wheal or hematoma formation, mild signs of pain at the injection site, and postvaccination bruising. CONCLUSIONS AND CLINICAL RELEVANCE: Results support the safety and efficacy of the huTyr DNA vaccine in dogs as adjunctive treatment for oral MM. IMPACT FOR HUMAN MEDICINE: Response to DNA vaccination in dogs with oral MM may be useful in development of plasmid DNA vaccination protocols for human patients with similar disease.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Enfermedades de los Perros/tratamiento farmacológico , Melanoma/veterinaria , Monofenol Monooxigenasa/uso terapéutico , Neoplasias de la Boca/veterinaria , Vacunas de ADN/uso terapéutico , Administración Cutánea , Animales , Vacunas contra el Cáncer/inmunología , ADN Complementario/genética , ADN Complementario/uso terapéutico , Enfermedades de los Perros/inmunología , Perros , Femenino , Estudios de Seguimiento , Humanos , Estimación de Kaplan-Meier , Masculino , Melanoma/tratamiento farmacológico , Melanoma/inmunología , Monofenol Monooxigenasa/genética , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/inmunología , Estadificación de Neoplasias/veterinaria , Procedimientos Quirúrgicos Orales/veterinaria , Plásmidos/genética , Plásmidos/uso terapéutico , Estudios Prospectivos , Resultado del Tratamiento , Estados Unidos , Vacunas de ADN/inmunología
11.
Exp Biol Med (Maywood) ; 236(10): 1211-7, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21885480

RESUMEN

Mutations in Pde6b lead to high levels of signaling molecules cyclic guanosine monophosphate (cGMP) and Ca(2+), which ultimately result in photoreceptor cell death in certain forms of retinitis pigmentosa (RP). The level of cGMP, which is controlled by opposing activities of guanylate cyclase (GUCY) and photoreceptor phosphodiesterase-6 (PDE6), regulates the opening of cyclic nucleotide-gated ion channels [CNG] and thereby controls Ca(2+) influx into the outer segments. Using a lentiviral gene therapy approach, we have previously shown that degeneration can be temporarily slowed either by introducing wild-type PDE6ß or knocking down expression of GUCY2E and CNGA1 in photoreceptors of Pde6b(H620Q), a mouse model for RP. Rescue was transient with either approach. Therefore, we tested a novel combination therapy using bipartite lentiviral vectors designed to both introduce wild-type PDE6ß expression and knockdown GUCY2E or CNGA1. Immunoblot analysis shows simultaneous increases in PDE6ß and decreases in GUCY2E or CNGA1 in retinas transduced by the vectors, indicating successful transduction. In Pde6b(H620Q) mutants, we observe rescue of photoreceptor function and an increase in photoreceptor rows as compared with untreated controls. However, no evidence of prolonged rescue beyond the limit of the previously tested single therapy was observed.


Asunto(s)
ADN Complementario/uso terapéutico , Terapia Genética/métodos , Lentivirus/genética , ARN Interferente Pequeño/uso terapéutico , Retinitis Pigmentosa/terapia , Animales , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/genética , Vectores Genéticos/genética , Vectores Genéticos/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Células Fotorreceptoras de Vertebrados/fisiología , Retina/metabolismo , Retina/virología , Retinitis Pigmentosa/genética , Transducción Genética/métodos
12.
J Clin Invest ; 121(6): 2160-8, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21606598

RESUMEN

Leber congenital amaurosis (LCA) is a rare degenerative eye disease, linked to mutations in at least 14 genes. A recent gene therapy trial in patients with LCA2, who have mutations in RPE65, demonstrated that subretinal injection of an adeno-associated virus (AAV) carrying the normal cDNA of that gene (AAV2-hRPE65v2) could markedly improve vision. However, it remains unclear how the visual cortex responds to recovery of retinal function after prolonged sensory deprivation. Here, 3 of the gene therapy trial subjects, treated at ages 8, 9, and 35 years, underwent functional MRI within 2 years of unilateral injection of AAV2-hRPE65v2. All subjects showed increased cortical activation in response to high- and medium-contrast stimuli after exposure to the treated compared with the untreated eye. Furthermore, we observed a correlation between the visual field maps and the distribution of cortical activations for the treated eyes. These data suggest that despite severe and long-term visual impairment, treated LCA2 patients have intact and responsive visual pathways. In addition, these data suggest that gene therapy resulted in not only sustained and improved visual ability, but also enhanced contrast sensitivity.


Asunto(s)
Proteínas Portadoras/fisiología , Proteínas del Ojo/fisiología , Terapia Genética , Amaurosis Congénita de Leber/terapia , Corteza Visual/fisiopatología , Adulto , Proteínas Portadoras/genética , Niño , ADN Complementario/administración & dosificación , ADN Complementario/genética , ADN Complementario/uso terapéutico , Dependovirus/genética , Proteínas del Ojo/genética , Vectores Genéticos/uso terapéutico , Humanos , Amaurosis Congénita de Leber/genética , Amaurosis Congénita de Leber/fisiopatología , Imagen por Resonancia Magnética , Estimulación Luminosa , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/fisiología , Recuperación de la Función , Reflejo Pupilar/efectos de la radiación , Privación Sensorial , Umbral Sensorial , cis-trans-Isomerasas
13.
Andrologia ; 41(3): 176-83, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19400852

RESUMEN

This work aimed to assess the efficacy of haeme oxygenase-1 (HO-1) cDNA-liposome complex transfer as a mediator of erectile signalling in aged rats. One hundred and fifty aged white albino rats were equally divided into five groups: controls, rats receiving lipofectamine, rats receiving intracorporeal HO-1 cDNA-lipsome complex, rats receiving HO-1 cDNA-liposome complex plus nitric oxide synthase (NOS) inhibitor, and rats receiving HO-1 cDNA-liposome complex plus HO inhibitor. Six rats were killed from each group after 12, 24 and 48 h, and after1 and 2 weeks. In dissected cavernous tissues, the following were assessed: HO-1 gene expression, Western blot for HO-1, HO enzyme activity, cGMP and histopathology. The results showed that HO-1 cDNA-liposome complex transfer led to a significant increase in cavernous tissue HO-1 protein, HO-1 gene expression, HO enzyme activity and cGMP up to 1 week. NOS inhibition exhibited no effect on HO-1 gene enhancement of cavernous tissue HO enzyme activity or cGMP, whereas inhibition of HO significantly decreased these parameters. Histopathology of cavernous tissue demonstrated a significant dilatation of helicine arteries in HO-1 cDNA-liposome complex treated group after 48 h compared with the controls. It is concluded that HO-1 cDNA-liposome complex transfer augments cavernous tissue cGMP with subsequent sinusoidal relaxation.


Asunto(s)
Disfunción Eréctil/terapia , Hemo-Oxigenasa 1/uso terapéutico , Liposomas/uso terapéutico , Erección Peniana/fisiología , Envejecimiento , Animales , Monóxido de Carbono/farmacología , ADN Complementario/uso terapéutico , Activación Enzimática/efectos de los fármacos , Expresión Génica , Técnicas de Transferencia de Gen , Guanilato Ciclasa/metabolismo , Hemo-Oxigenasa 1/biosíntesis , Masculino , NG-Nitroarginina Metil Éster/uso terapéutico , Erección Peniana/genética , Ratas , Receptores Citoplasmáticos y Nucleares/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Guanilil Ciclasa Soluble
14.
Spine (Phila Pa 1976) ; 33(14): 1509-16; discussion 1517, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18520636

RESUMEN

STUDY DESIGN: Clinical, biochemical, and histologic analysis was performed after in vivo delivery of cDNA encoding various anabolic cytokines and marker genes to the lumbar epidural space of New Zealand white rabbits, using both adenoviral and adeno-associated viral vectors. OBJECTIVE: To mimic errant or misplaced doses of gene therapy to better ascertain the potential risks associated with alternative vectors and transgene products with regard to their application to problems of the intervertebral disc. SUMMARY OF BACKGROUND DATA: Work done with several anabolic cytokines including bone morphogenic proteins and transforming growth factors, has demonstrated the potential of gene therapy. Recently, data has been published demonstrating that improperly dosed or delivered adenoviral-mediated gene therapy within the subarachnoid space can result in significant morbidity in rabbits. There are currently no studies examining the effect of these errors within the epidural space or using an adeno-associated viral (AAV) vector. METHODS: Using either adenoviral or AAV vectors, complementary DNA (cDNA) encoding anabolic cytokines bone morphogenic protein-2 (BMP-2) and transforming growth factor-beta 1 and marker proteins LacZ and green fluorescent protein were injected into the epidural space of 37 New Zealand white rabbits at the L5/6 level. Rabbits were then observed clinically for up to 6 weeks, after which the rabbits were sacrificed in order to perform a comprehensive biochemical and histologic analysis. RESULTS: Following adenoviral-mediated delivery of anabolic cytokine cDNA, up to eighty percent of rabbits demonstrated significant clinical, biochemical, and histologic morbidity. Conversely, AAV-mediated delivery of any cDNA and adenoviral-mediated delivery of marker protein cDNA resulted in no clinical, histologic, or biochemical morbidity. CONCLUSION: Properly dosed and directed gene therapy seems to be both safe and potentially efficacious. This study suggests that side effects of gene therapy may be due to a combination of dosing, transgene product, and vector choice, and that newer AAV vectors may reduce these side-effects and decrease the risk of this technology.


Asunto(s)
Adenoviridae/genética , ADN Complementario/uso terapéutico , Dependovirus/genética , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos , Enfermedades de la Columna Vertebral/terapia , Animales , Proteína Morfogenética Ósea 2 , Proteínas Morfogenéticas Óseas/genética , ADN Complementario/administración & dosificación , Modelos Animales de Enfermedad , Espacio Epidural , Femenino , Proteínas Fluorescentes Verdes/genética , Inyecciones Espinales , Operón Lac/genética , Vértebras Lumbares , Conejos , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta1/genética
15.
Arthritis Rheum ; 58(4): 1026-37, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18383372

RESUMEN

OBJECTIVE: Vasoactive intestinal peptide (VIP) has been shown to exert potent immunomodulatory activity, and the use of lentiviral vectors has been found to be an effective means of gene delivery. The present study was therefore undertaken to investigate the feasibility and efficiency of gene therapy using lentiviral vectors expressing VIP (LentiVIP) for the treatment of rheumatoid arthritis (RA). METHODS: We evaluated the therapeutic potential of the gene therapy strategy in the collagen-induced arthritis (CIA) mouse model, administering the vectors at different phases of the disease. The inflammatory response was determined by measuring the levels of various inflammatory cytokines and chemokines in the joints and serum. The Th1-mediated response was evaluated by determining the proliferative response and cytokine profile of T cells stimulated with autoantigen. RESULTS: A single intraperitoneal injection of LentiVIP was highly effective in treating CIA. Mice with established, severe arthritis showed complete regression of the disease. The therapeutic effect of LentiVIP was associated with widespread biodistribution of the vector and increased VIP levels, especially in joints and lymphoid organs, and was mediated through a striking reduction of the 2 deleterious components of the disease, i.e., the autoimmune response (self-reactive Th1 cell activity and autoantibody production) and the inflammatory response. LentiVIP treatment also induced the generation and/or activation of CD4+,CD25+,FoxP3+ Treg cells in arthritic mice. CONCLUSION: Our findings show that in vivo administration of lentiviral vector expressing VIP produces one of the most potent therapeutic effects described so far in any animal model of RA. We propose that VIP gene transfer should be further investigated as a potential novel, effective treatment of RA and other chronic autoimmune disorders.


Asunto(s)
Artritis Experimental/genética , Artritis Experimental/terapia , ADN Complementario/uso terapéutico , Terapia Genética , Péptido Intestinal Vasoactivo/genética , Animales , Artritis Experimental/metabolismo , Vectores Genéticos/uso terapéutico , Inflamación/terapia , Lentivirus/genética , Ratones , Linfocitos T Reguladores/inmunología , Células TH1/efectos de los fármacos , Células TH1/inmunología , Péptido Intestinal Vasoactivo/uso terapéutico
16.
FASEB J ; 21(9): 2135-48, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17341681

RESUMEN

Immunotherapy with Abeta is expected to bring great improvement for Alzheimer disease (AD). However, clinical trials have been suspended because of meningoencephalitics, which accompanied lymphocytic infiltration. We have developed an oral vaccine for AD with a recombinant adeno-associated viral vector carrying Abeta cDNA (AAV/Abeta). The vaccine reduces the amount of Abeta deposited without lymphocytic infiltration in APP transgenic (Tg2576) mice. In the present study, Tg2576 mice showed progressive cognitive impairments in the novel object recognition test, Y-maze test, water maze test, and contextual conditioned fear learning test. A single oral administration of AAV/Abeta to Tg2576 mice at the age of 10 months alleviated progressive cognitive impairment with decreased Abeta deposition, insoluble Abeta, soluble Abeta oligomer (Abeta*56), microglial attraction, and synaptic degeneration induced in the brain regions at the age of 13 months. A histological analysis with hematoxylin and eosin and an immunohistochemical analysis with antibodies against CD3, CD4, CD8, and CD19 suggested there was no lymphocytic infiltration or microhemorrhage in the brain of AAV/Abeta-vaccinated Tg2576 mice at 13 months of age. Taken together, these results suggest that immunotherapy with AAV/Abeta is a safe and effective treatment for AD.


Asunto(s)
Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/inmunología , Vectores Genéticos/uso terapéutico , Inmunoterapia Activa , Fragmentos de Péptidos/inmunología , Vacunación , Vacunas de ADN/uso terapéutico , Administración Oral , Enfermedad de Alzheimer/psicología , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/genética , Animales , Aprendizaje por Asociación , Reacción de Prevención , Encéfalo/inmunología , Encéfalo/patología , Química Encefálica , ADN Complementario/genética , ADN Complementario/inmunología , ADN Complementario/uso terapéutico , Dependovirus/genética , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Conducta Exploratoria , Miedo , Femenino , Reacción Cataléptica de Congelación , Vectores Genéticos/inmunología , Aprendizaje por Laberinto , Ratones , Ratones Transgénicos , Microglía/patología , Actividad Motora , Mutación Missense , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Placa Amiloide , Mutación Puntual , Reconocimiento en Psicología , Solubilidad , Sinapsis/patología , Vacunas de ADN/inmunología
17.
Med Sci (Paris) ; 23(3): 303-9, 2007 Mar.
Artículo en Francés | MEDLINE | ID: mdl-17349293

RESUMEN

Osteoarticular disorders are the major cause of disability in Europe and North America. It is estimated that rheumatoid arthritis affects 1 % of the population and that more than two third of people over age 55 develop osteoarthritis. Because there are no satisfactory treatments, gene therapy offers a new therapeutic approach. The delivery of cDNA encoding anti-arthritic proteins to articular cells has shown therapeutic efficacy in numerous animal models in vivo. Through the development and the experimental progresses that have been made for both rheumatoid arthritis and osteoarthritis, this review discusses the different gene therapy strategies available today and the safety issues with which they may be associated. Among the different vectors available today, adeno-associated virus seems the best candidate for a direct in vivo gene delivery approach for the treatment of joint disorders.


Asunto(s)
Artritis Reumatoide/terapia , Terapia Genética , Osteoartritis/terapia , Anciano , Animales , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/fisiopatología , Cartílago Articular/metabolismo , Cartílago Articular/patología , Citocinas/antagonistas & inhibidores , Citocinas/genética , ADN Complementario/administración & dosificación , ADN Complementario/uso terapéutico , Dependovirus/genética , Perros , Doxiciclina/farmacología , Etanercept , Expresión Génica/efectos de los fármacos , Genes Sintéticos , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/efectos adversos , Vectores Genéticos/uso terapéutico , Haplorrinos , Caballos , Humanos , Inmunoglobulina G/uso terapéutico , Inyecciones Intraarticulares , Ratones , Persona de Mediana Edad , Osteoartritis/fisiopatología , Receptores del Factor de Necrosis Tumoral/uso terapéutico , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Sirolimus/farmacología
19.
DNA Cell Biol ; 24(11): 751-7, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16274295

RESUMEN

The objective of this study was to investigate the efficacy of combination gene therapy with multiple angiogenic growth factor cDNAs to enhance survival of ischemic skin flaps in a rat model. Sixty Sprague-Dawley rats were divided into six groups. Varying combinations of VEGF165, PDGF-B, and bFGF-plasmids were injected to prefabricate the flaps. Random skin flaps were raised on the dorsal aspect of rats following prefabrication with growth factor cDNAs. Flap viability was determined by measurement of percentage area of survival. The efficacy of gene therapy was evaluated by flap survival and neovascularization of representative histologic sections stained immunohistologically. The VEGF165 plus bFGF cDNAs enhanced the viability of the flap and neovascularization most effectively; the flap survival area was 64.3 +/- 8.7% after transfer of these two growth factor genes. Addition of PDGF-B cDNA is deleterious to the effects of combined VEGF165 and bFGF, leading to a significant decrease in flap viability (44.9 +/- 2.7%). Viability of the flaps with combined VEGF165 and bFGF cDNA transfer was significantly greater than that of the flaps with VEGF165 transfer alone (57.6 +/- 5.2%) or sham plasmid control (52.3 +/- 5.0%). Combined transfer of VEGF165 and bFGF cDNA is the most effective combination of multiple growth factor genes to improve flap viability in this model. Simultaneous transfer of three growth factor genes (VEGF165, PDGF-B, and bFGF) is deleterious to flap survival, at least for the ratio of lipofectin:transgene employed.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/genética , Terapia Genética , Neovascularización Fisiológica , Factor de Crecimiento Derivado de Plaquetas/genética , Piel/irrigación sanguínea , Colgajos Quirúrgicos/irrigación sanguínea , Factor A de Crecimiento Endotelial Vascular/genética , Cicatrización de Heridas , Animales , Supervivencia Celular , Terapia Combinada , ADN Complementario/uso terapéutico , Femenino , Plásmidos , Ratas , Ratas Sprague-Dawley
20.
J Immunol ; 175(6): 3900-6, 2005 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16148136

RESUMEN

Coccidioides posadasii causes coccidioidomycosis, or Valley fever, in the endemic regions of the Southwestern United States. The susceptibility to C. posadasii infection has been attributed to a decreased Th1 cellular response. APCs, especially dendritic cells (DCs), play an important role in the activation of Th1 response. In this study, we investigated the efficacy of a DC-based vaccine against C. posadasii in a mouse model of coccidioidomycosis. We intranasally immunized C57BL6 mice with syngeneic, bone marrow-derived DCs (JAWS II cells) transfected with a cDNA encoding the protective Coccidioides-Ag2/proline-rich Ag. The immunized mice were lethally challenged with C. posadasii through either an i.p. or intranasal route. Upon necropsy after 10 days of infection, fungal burden in lung and spleen of immunized mice was significantly reduced as compared with the control animals. The lung tissue homogenates of immunized animals showed higher levels of IFN-gamma. Histologically, lung tissues of immunized mice were in better condition than the control mice. To further investigate, we studied the biodistribution and trafficking of injected DCs by nuclear imaging techniques. For this purpose, the transfected DCs were radiolabeled with (111)In-oxime. Scintigraphic images showed that most of the label remained in the gastrointestinal tract. A significant amount was also observed in lung, but there were negligible circulating (111)In label in blood. The results suggest that the DCs have a potent immunostimulatory activity, and immunization with DCs transfected with Ag2/proline-rich Ag-cDNA induces protective immunity against C. posadasii in C57BL6 mice.


Asunto(s)
Coccidioidomicosis/terapia , Células Dendríticas/inmunología , Células Dendríticas/trasplante , Proteínas Fúngicas/genética , Glicoproteínas/genética , Inmunización/métodos , Traslado Adoptivo/métodos , Animales , Coccidioides/inmunología , ADN Complementario/uso terapéutico , Células Dendríticas/metabolismo , Proteínas Fúngicas/uso terapéutico , Glicoproteínas/uso terapéutico , Radioisótopos de Indio/farmacocinética , Interferón gamma/análisis , Pulmón/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Cintigrafía , Bazo/microbiología , Células TH1 , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA