Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mov Disord Clin Pract ; 7(1): 70-77, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31970214

RESUMEN

BACKGROUND: Adenylate cyclase 5 (ADCY5)-related dyskinesia is a childhood-onset movement disorder. Manifestations vary in frequency and severity and may include chorea, tremor, dystonia, facial twitches, myoclonus, axial hypotonia, and limb hypertonia. Psychosis is likely part of the broader spectrum. ADCY5 is widely expressed in the brain, especially in the striatum. Previous reports of brain autopsies of 2 subjects with likely ADCY5-dyskinesia were limited by the absence of a molecular diagnosis. In 1 case, normal gross pathology was reported. In the other case, ADCY5 expression was not examined and neuropathological findings were confounded by age and comorbidities. OBJECTIVES: To examine ADCY5 expression and neuropathological changes in ADCY5-dyskinesia. METHODS: An extensive brain autopsy, including immunohistochemical analyses with antibodies to paired helical filament tau, α-synuclein, amyloid-ß, microtubule-associated protein 2, and ADCY5, was performed. RESULTS: The patient, with a p.M1029K ADCY5 variant, had severe dyskinesias from early childhood, later recurrent episodes of psychosis, and died at age 46. Gross pathology was unremarkable, but we detected increased immunoreactivity for ADCY5 in neurons in multiple brain regions. Despite no history of brain trauma to suggest chronic traumatic encephalopathy, we found tau deposits in the deep cortical sulci, midbrain, and hippocampus with minimal amyloid pathology and no Lewy bodies. CONCLUSIONS: We present the first brain autopsy findings in a molecularly proven case of ADCY5-dyskinesia, showing increased ADCY5 immunoreactivity in neurons and evidence of tau deposition. Additional patients will need to be studied to determine whether increased immunoreactivity for ADCY5 is a signature for ADCY5-dyskinesia and whether this disease has a tauopathy component.

2.
Muscle Nerve ; 57(5): 859-862, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29236290

RESUMEN

INTRODUCTION: Mutations in gap junction protein beta 1 (GJB1) on the X chromosome represent one of the most common causes of hereditary neuropathy. We assessed manifestations associated with a rare 3' untranslated region mutation (UTR) of GJB1 in a large family with X-linked Charcot-Marie-Tooth disease (CMTX). METHODS: Clinical, electrophysiological, and molecular genetic analyses were performed on an 8-generation family with CMTX. RESULTS: There were 22 affected males and 19 symptomatic females, including an 83-year-old woman followed for 40 years. Electrophysiological studies showed a primarily axonal neuropathy. The c.*15C>T mutation in the GJB1 3' UTR was identified in 4 branches of the family with a log of odds (LOD) of 4.91. This created a BstE II enzyme recognition site that enabled detection by restriction digestion. DISCUSSION: The c.*15C>T mutation in the GJB1 3' UTR segregates with CMTX1 in 8 generations. Penetrance in males and females is essentially complete. A straightforward genetic method to detect this mutation is described. Muscle Nerve 57: 859-862, 2018.


Asunto(s)
Regiones no Traducidas 3'/genética , Enfermedad de Charcot-Marie-Tooth/genética , Conexinas/genética , Salud de la Familia , Mutación/genética , Adolescente , Adulto , Enfermedad de Charcot-Marie-Tooth/fisiopatología , Niño , Femenino , Perfilación de la Expresión Génica , Pruebas Genéticas , Genotipo , Humanos , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteína beta1 de Unión Comunicante
3.
Bioorg Med Chem Lett ; 25(10): 2133-40, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25891106

RESUMEN

Noscapine is an opium-derived kinder-gentler microtubule-modulating drug, currently in Phase I/II clinical trials for cancer chemotherapy. Here, we report the synthesis of four more potent di-substituted brominated derivatives of noscapine, 9-Br-7-OH-NOS (2), 9-Br-7-OCONHEt-NOS (3), 9-Br-7-OCONHBn-NOS (4), and 9-Br-7-OAc-NOS (5) and their chemotherapeutic efficacy on PC-3 and MDA-MB-231 cells. The four derivatives were observed to have higher tubulin binding activity than noscapine and significantly affect tubulin polymerization. The equilibrium dissociation constant (KD) for the interaction between tubulin and 2, 3, 4, 5 was found to be, 55±6µM, 44±6µM, 26±3µM, and 21±1µM respectively, which is comparable to parent analog. The effects of these di-substituted noscapine analogs on cell cycle parameters indicate that the cells enter a quiescent phase without undergoing further cell division. The varying biological activity of these analogs and bulk of substituent at position-7 of the benzofuranone ring system of the parent molecule was rationalized utilizing predictive in silico molecular modeling. Furthermore, the immunoblot analysis of protein lysates from cells treated with 4 and 5, revealed the induction of apoptosis and down-regulation of survivin levels. This result was further supported by the enhanced activity of caspase-3/7 enzymes in treated samples compared to the controls. Hence, these compounds showed a great potential for studying microtubule-mediated processes and as chemotherapeutic agents for the management of human cancers.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Diseño de Fármacos , Microtúbulos/efectos de los fármacos , Noscapina/síntesis química , Noscapina/farmacología , Antineoplásicos/química , Línea Celular Tumoral , Humanos , Modelos Moleculares , Noscapina/química
4.
J Immunol ; 191(6): 3358-72, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23956422

RESUMEN

Recent reports have highlighted greater complexity, plasticity, and functional diversity of mononuclear phagocytes (MPCs), including monocytes, macrophages, and dendritic cells (DCs), in our organs than previously understood. The functions and origins of MPCs resident within healthy organs, especially in the kidney, are less well understood, whereas studies suggest they play roles in disease states distinct from recruited monocytes. We developed an unbiased approach using flow cytometry to analyze MPCs residing in the normal mouse kidney, and identified five discrete subpopulations according to CD11b/CD11c expression as well as F4/80, CD103, CD14, CD16, and CD64 expression. In addition to distinct marker profiles, these subpopulations have different lineages and expression of genes involved in tissue homeostasis, including angiogenesis. Among them, the CD11b(int)CD11c(int) F4/80(high) subpopulation notably exhibited high capacity to produce a representative anti-inflammatory cytokine, IL-10. Each subpopulation had different degrees of both macrophage (phagocytosis) and DC (Ag presentation) capacities, with a tendency to promote differentiation of regulatory T cells, whereas two of these showed expression of transcription factors reported to be highly expressed by classical DCs, and proclivity to exit the kidney following stimulation with LPS. In summary, resident kidney MPCs comprise discrete subpopulations, which cannot be simply classified into the conventional entities, and they produce anti-inflammatory and tissue-homeostatic factors to differing degrees.


Asunto(s)
Riñón/citología , Riñón/inmunología , Sistema Mononuclear Fagocítico/citología , Sistema Mononuclear Fagocítico/inmunología , Animales , Diferenciación Celular/inmunología , Citometría de Flujo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microscopía Fluorescente , Sistema Mononuclear Fagocítico/metabolismo , Fenotipo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/citología , Linfocitos T/inmunología
5.
Small ; 9(11): 1964-73, 2013 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-23292656

RESUMEN

Molecular therapy using a small interfering RNA (siRNA) has shown promise in the development of novel therapeutics. Various formulations have been used for in vivo delivery of siRNAs. However, the stability of short double-stranded RNA molecules in the blood and efficiency of siRNA delivery into target organs or tissues following systemic administration have been the major issues that limit applications of siRNA in human patients. In this study, multifunctional siRNA delivery nanoparticles are developed that combine imaging capability of nanoparticles with urokinase plasminogen activator receptor-targeted delivery of siRNA expressing DNA nanocassettes. This theranostic nanoparticle platform consists of a nanoparticle conjugated with targeting ligands and double-stranded DNA nanocassettes containing a U6 promoter and a shRNA gene for in vivo siRNA expression. Targeted delivery and gene silencing efficiency of firefly luciferase siRNA nanogenerators are demonstrated in tumor cells and in animal tumor models. Delivery of survivin siRNA expressing nanocassettes into tumor cells induces apoptotic cell death and sensitizes cells to chemotherapy drugs. The ability of expression of siRNAs from multiple nanocassettes conjugated to a single nanoparticle following receptor-mediated internalization should enhance the therapeutic effect of the siRNA-mediated cancer therapy.


Asunto(s)
Nanopartículas/química , ARN Interferente Pequeño/genética , Línea Celular Tumoral , Silenciador del Gen , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Survivin
6.
Carcinogenesis ; 33(9): 1769-81, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22678119

RESUMEN

Endothelial tubular morphogenesis relies on an exquisite interplay of microtubule dynamics and actin remodeling to propel directed cell migration. Recently, the dynamicity and integrity of microtubules have been implicated in the trafficking and efficient translation of the mRNA for HIF-1α (hypoxia-inducible factor), the master regulator of tumor angiogenesis. Thus, microtubule-disrupting agents that perturb the HIF-1α axis and neovascularization cascade are attractive anticancer drug candidates. Here we show that EM011 (9-bromonoscapine), a microtubule-modulating agent, inhibits a spectrum of angiogenic events by interfering with endothelial cell invasion, migration and proliferation. Employing green-fluorescent transgenic zebrafish, we found that EM011 not only inhibited vasculogenesis but also disrupted preexisting vasculature. Mechanistically, EM011 caused proteasome-dependent, VHL-independent HIF-1α degradation and repressed expression of HIF-1α downstream targets, namely VEGF and survivin. Furthermore, EM011 inhibited membrane ruffling and impeded formation of filopodia, lamellipodia and stress fibers, which are critical for cell migration. These events were associated with a drug-mediated decrease in activation of Rho GTPases- RhoA, Cdc42 and Rac1, and correlated with a loss in the geometric precision of centrosome reorientation in the direction of movement. This is the first report to describe a previously unrecognized, antiangiogenic property of a noscapinoid, EM011, and provides evidence for novel anticancer strategies recruited by microtubule-modulating drugs.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Dioxoles/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Isoquinolinas/farmacología , Animales , Movimiento Celular/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Células Cultivadas , Centrosoma/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Humanos , Masculino , Ratones , Microtúbulos/efectos de los fármacos , Paxillin/fisiología , Activación Transcripcional , Proteínas de Unión al GTP rho/metabolismo
7.
Lab Invest ; 92(4): 499-510, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22249313

RESUMEN

Glomerular parietal epithelial cells (PECs) are precursors to podocytes in mature glomeruli; however, as progenitors, the distinct intrinsic mechanisms that allow for repeated periods of cell-cycle arrest and re-entry of PECs after glomerulogenesis are unknown. Here, we show that the Src-suppressed protein kinase C substrate (SSeCKS), a multivalent scaffolding A kinase anchoring protein, sequesters cyclin D1 in the cytoplasm of quiescent PECs. SSeCKS expression is induced in embryonic PECs, but not in embryonic podocytes, starting at the S phase of glomerulogenesis, and is constitutively expressed postnatally by PECs, but not podocytes, in normal glomeruli. Cyclin D1 was immunoprecipitated with SSeCKS from capsulated glomeruli containing PECs, whereas decapsulated glomeruli without PECs lacked SSeCKS and cyclin D1. Cell-cell contact inhibition of proliferation in cultured PECs induced SSeCKS expression and binding of cyclin D1 by SSeCKS in the cytoplasm, whereas phosphorylation of SSeCKS by activated protein kinase C disrupted binding, resulting in nuclear translocation of cyclin D1. SSeCKS(-/-) mice showed hyperplasia of PECs in otherwise normal glomeruli and developed significantly worse proteinuric glomerular disease, marked by increased PEC proliferation and expression of nuclear cyclin D1, from nephrotoxic nephritis. These results suggest that SSeCKS controls the localization and activity of cyclin D1 in PECs and influences proliferative injury in the glomerulus.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ciclina D1/metabolismo , Glomérulos Renales/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Glomérulos Renales/embriología , Masculino , Ratones , Ratones Noqueados , Fenotipo , Podocitos/metabolismo
8.
Br J Nutr ; 107(4): 473-84, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21849094

RESUMEN

It is appreciated far and wide that increased and regular consumption of fruits and vegetables is linked with noteworthy anticancer benefits. Extensively consumed as a spice in foods and beverages worldwide, ginger (Zingiber officinale Roscoe) is an excellent source of several bioactive phenolics, including non-volatile pungent compounds such as gingerols, paradols, shogaols and gingerones. Ginger has been known to display anti-inflammatory, antioxidant and antiproliferative activities, indicating its promising role as a chemopreventive agent. Here, we show that whole ginger extract (GE) exerts significant growth-inhibitory and death-inductory effects in a spectrum of prostate cancer cells. Comprehensive studies have confirmed that GE perturbed cell-cycle progression, impaired reproductive capacity, modulated cell-cycle and apoptosis regulatory molecules and induced a caspase-driven, mitochondrially mediated apoptosis in human prostate cancer cells. Remarkably, daily oral feeding of 100 mg/kg body weight of GE inhibited growth and progression of PC-3 xenografts by approximately 56 % in nude mice, as shown by measurements of tumour volume. Tumour tissue from GE-treated mice showed reduced proliferation index and widespread apoptosis compared with controls, as determined by immunoblotting and immunohistochemical methods. Most importantly, GE did not exert any detectable toxicity in normal, rapidly dividing tissues such as gut and bone marrow. To the best of our knowledge, this is the first report to demonstrate the in vitro and in vivo anticancer activity of whole GE for the management of prostate cancer.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Suplementos Dietéticos , Extractos Vegetales/uso terapéutico , Neoplasias de la Próstata/dietoterapia , Rizoma/química , Zingiber officinale/química , Animales , Antineoplásicos Fitogénicos/efectos adversos , Antineoplásicos Fitogénicos/metabolismo , Apoptosis , Proteínas Reguladoras de la Apoptosis/metabolismo , Biomarcadores/metabolismo , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Suplementos Dietéticos/efectos adversos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Extractos Vegetales/efectos adversos , Extractos Vegetales/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/prevención & control , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Biomaterials ; 2011 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-22169827

RESUMEN

This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.

10.
Carcinogenesis ; 32(12): 1872-80, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21948980

RESUMEN

Sweet potato (Ipomoea batatas) leaves or greens, extensively consumed as a vegetable in Africa and Asia, are an excellent source of dietary polyphenols such as anthocyanins and phenolic acids. Here, we show that sweet potato greens extract (SPGE) has the maximum polyphenol content compared with several commercial vegetables including spinach. The polyphenol-rich SPGE exerts significant antiproliferative activity in a panel of prostate cancer cell lines while sparing normal prostate epithelial cells. Mechanistically, SPGE perturbed cell cycle progression, reduced clonogenic survival, modulated cell cycle and apoptosis regulatory molecules and induced apoptosis in human prostate cancer PC-3 cells both in vitro and in vivo. SPGE-induced apoptosis has a mitochondrially mediated component, which was attenuated by pretreatment with cyclosporin A. We also observed alterations of apoptosis regulatory molecules such as inactivation of Bcl2, upregulation of BAX, cytochrome c release and activation of downstream apoptotic signaling. SPGE caused DNA degradation as evident by terminal deoxynucleotidyl transferase-mediated dUTP-nick-end labeling (TUNEL) staining of increased concentration of 3'-DNA ends. Furthermore, apoptotic induction was caspase dependent as shown by cleavage of caspase substrate, poly (adenosine diphosphate-ribose) polymerase. Oral administration of 400 mg/kg SPGE remarkably inhibited growth and progression of prostate tumor xenografts by ∼69% in nude mice, as shown by tumor volume measurements and non-invasive real-time bioluminescent imaging. Most importantly, SPGE did not cause any detectable toxicity to rapidly dividing normal tissues such as gut and bone marrow. This is the first report to demonstrate the in vitro and in vivo anticancer activity of sweet potato greens in prostate cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ipomoea batatas/química , Hojas de la Planta/química , Polifenoles/farmacología , Neoplasias de la Próstata/patología , Animales , Línea Celular Tumoral , Humanos , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Ratones Desnudos , Microscopía Fluorescente
11.
Biochem Pharmacol ; 82(2): 110-21, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21501599

RESUMEN

Microtubules, composed of α/ß tubulin heterodimers, represent a validated target for cancer chemotherapy. Thus, tubulin- and microtubule-binding antimitotic drugs such as taxanes and vincas are widely employed for the chemotherapeutic management of various malignancies. Although quite successful in the clinic, these drugs are associated with severe toxicity and drug resistance problems. Noscapinoids represent an emerging class of microtubule-modulating anticancer agents based upon the parent molecule noscapine, a naturally occurring non-toxic cough-suppressant opium alkaloid. Here we report in silico molecular modeling, chemical synthesis and biological evaluation of novel analogs derived by modification at position-7 of the benzofuranone ring system of noscapine. The synthesized analogs were evaluated for their tubulin polymerization activity and their biological activity was examined by their antiproliferative potential using representative cancer cell lines from varying tissue-origin [A549 (lung), CEM (lymphoma), MIA PaCa-2 (pancreatic), MCF-7 (breast) and PC-3 (prostate)]. Cell-cycle studies were performed to explore their ability to halt the cell-cycle and induce subsequent apoptosis. The varying biological activity of these analogs that differ in the nature and bulk of substituent at position-7 was rationalized utilizing predictive in silico molecular modeling.


Asunto(s)
Antineoplásicos/síntesis química , Benzofuranos/síntesis química , Noscapina/análogos & derivados , Moduladores de Tubulina/síntesis química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Benzofuranos/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Simulación por Computador , Humanos , Modelos Moleculares , Noscapina/síntesis química , Noscapina/farmacología , Estructura Cuaternaria de Proteína , Relación Estructura-Actividad , Tubulina (Proteína)/química , Moduladores de Tubulina/farmacología
12.
J Biol Chem ; 286(11): 9280-6, 2011 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-21220424

RESUMEN

Reversible acetylation of Tat is critical for its transactivation activity toward HIV-1 transcription. However, the enzymes involved in the acetylation/deacetylation cycles have not been fully characterized. In this study, by yeast two-hybrid assay, we have discovered the histone deacetylase HDAC6 to be a binding partner of Tat. Our data show that HDAC6 interacts with Tat in the cytoplasm in a microtubule-dependent manner. In addition, HDAC6 deacetylates Tat at Lys-28 and thereby suppresses Tat-mediated transactivation of the HIV-1 promoter. Inactivation of HDAC6 promotes the interaction of Tat with cyclin T1 and leads to an increase in Tat transactivation activity. These findings establish HDAC6 as a Tat deacetylase and support a model in which Lys-28 deacetylation decreases Tat transactivation activity through affecting the ability of Tat to form a ribonucleoprotein complex with cyclin T1 and the transactivation-responsive RNA.


Asunto(s)
Regulación Viral de la Expresión Génica/fisiología , VIH-1/metabolismo , Histona Desacetilasas/metabolismo , Transcripción Genética/fisiología , Activación Transcripcional/fisiología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Acetilación , Animales , Ciclina T/genética , Ciclina T/metabolismo , Células HEK293 , VIH-1/genética , Histona Desacetilasa 6 , Histona Desacetilasas/genética , Humanos , Células Jurkat , Ratones , Ratones Noqueados , Microtúbulos/genética , Microtúbulos/metabolismo , Regiones Promotoras Genéticas/fisiología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
13.
Biochem Pharmacol ; 81(4): 478-87, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21087597

RESUMEN

Prostate cancer has been widely viewed as a chemoresistant neoplasm. Perhaps, the most prevalent antimicrotubule strategy involves docetaxel administration at its maximum-tolerated dose (MTD). Although the goal is to obtain total eradication of cancer cells, debilitating toxicities are presented by docetaxel therapy, including myelosuppression, immunosuppression, gastrointestinal toxicity and peripheral neuropathy. In addition, solubility limitations necessitate infusion of high-doses intravenously once or twice a week followed by a rest period, which allows recovery of normal proliferating cells to counter-balance efficacy. An emerging notion is that more of a toxic drug at its MTD is not necessarily better. It is likely that combinatorial antimicrotubule therapy with drugs occupying different sites on tubulin may enhance efficacy while reducing toxicity. Here we show that bromonoscapine (EM011), a microtubule-modulating noscapine analog, displays synergism with docetaxel as seen by cell viability and proliferation assays. Cell-cycle data demonstrated that lower dose-levels of docetaxel (25nM) in combination with EM011 caused an additive increase in proapoptotic activity. Since docetaxel alone caused severe mitotic arrest followed by mitotic slippage and endoreduplication, we strategized a sequential treatment regime that involved initial pretreatment with docetaxel followed by addition of EM011 to maximize mitotic arrest and subsequent apoptosis. In vivo studies with docetaxel and EM011 in combination showed a marked inhibition of tumor growth compared to docetaxel or EM011 as single-agents. Our studies suggest the potential usefulness of EM011 in the clinic to enhance docetaxel activity. This would reduce toxicity, thus improving the quality of life of docetaxel-treated patients.


Asunto(s)
Neoplasias de la Próstata/tratamiento farmacológico , Moduladores de Tubulina/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Dioxoles/uso terapéutico , Docetaxel , Humanos , Isoquinolinas/uso terapéutico , Masculino , Taxoides/uso terapéutico
14.
J Control Release ; 149(3): 314-22, 2011 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-21047537

RESUMEN

The tubulin-binding anticancer activity of noscapine, an orally available plant-derived anti-tussive alkaloid, has been recently identified. Noscapine inhibits tumor growth in nude mice bearing human xenografts of hematopoietic, breast, lung, ovarian, brain and prostate origin. Despite its nontoxic attributes, significant elimination of the disease has not been achieved, perhaps since the bioavailability of noscapine to tumors saturates at an oral dose of 300 mg/kg body weight. To enable the selective and specific delivery of noscapine to prostate cancer cells, we have engineered a multifunctional nanoscale delivery vehicle that takes advantage of urokinase plasminogen activator receptor (uPAR) overexpression in prostate cancer compared to normal prostate epithelia and can be tracked by magnetic resonance imaging (MRI) and near-infrared (NIR) imaging. Specifically, we employed the human-type 135 amino-acid amino-terminal fragment (hATF) of urokinase plasminogen activator (uPA), a high-affinity natural ligand for uPAR. Noscapine (Nos) was efficiently adsorbed onto the amphiphilic polymer coating of uPAR-targeted nanoparticles (NPs). Nos-loaded NPs were uniformly compact-sized, stable at physiological pH and efficiently released the drug at pH 4 to 5 within a span of 4h. Our results demonstrate that these uPAR-targeted NPs were capable of binding to the receptor and were internalized by PC-3 cells. uPAR-targeted Nos-loaded NPs enhanced intracellular noscapine accumulation as evident by the ~6-fold stronger inhibitory effect on PC-3 growth compared to free noscapine. In addition, Nos-loaded iron oxide NPs maintained their T2 MRI contrast effect upon internalization into tumor cells owing to their significant susceptibility effect in cells. Thus, our data provide compelling evidence that these optically and magnetic resonance imaging (MRI)-trackable uPAR-targeted NPs may offer a great potential for image-directed targeted delivery of noscapine for the management of prostate cancer.


Asunto(s)
Antineoplásicos/administración & dosificación , Antitusígenos/administración & dosificación , Imagen por Resonancia Magnética/métodos , Nanopartículas/química , Noscapina/administración & dosificación , Neoplasias de la Próstata/tratamiento farmacológico , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Antitusígenos/farmacocinética , Antitusígenos/farmacología , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Nanopartículas/ultraestructura , Noscapina/farmacocinética , Noscapina/farmacología , Próstata/efectos de los fármacos , Próstata/patología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética
15.
J Biol Chem ; 285(24): 18737-48, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20404319

RESUMEN

Autophagy is being increasingly implicated in both cell survival and death. However, the intricate relationships between drug-induced autophagy and apoptosis remain elusive. Here we demonstrate that a tubulin-binding noscapine analog, (R)-9-bromo-5-((S)-4,5-dimethoxy-1,3-dihydroisobenzofuran-1-yl)-4-methoxy-6-methyl-5,6,7,8-tetrahydro-[1,3]-di-oxolo[4,5-g]isoquinoline (Red-Br-nos), exerts a novel autophagic response followed by apoptotic cell death in human prostate cancer PC-3 cells. Red-Br-nos-induced autophagy was an early event detectable within 12 h that displayed a wide array of characteristic features including double membranous vacuoles with entrapped organelles, acidic vesicular organelles, and increased expression of LC3-II and beclin-1. Red-Br-nos-triggered release of reactive oxygen species (ROS) and attenuation of ROS by tiron, a ROS scavenger, reduced the sub-G(1) population suggesting ROS-dependent apoptosis. Abrogation of ROS also reduced autophagy indicating that ROS triggers autophagy. Pharmacological and genetic approaches to inhibit autophagy uncovered the protective role of Red-Br-nos-induced autophagy in PC-3 cells. Direct effects of the drug on mitochondria viz. disruption of normal cristae architecture and dissipation of mitochondrial transmembrane potential revealed a functional link between ROS generation, autophagy, and apoptosis induction. This is the first report to demonstrate the protective role of ROS-mediated autophagy and induction of caspase-independent ROS-dependent apoptosis in PC-3 cells by Red-Br-nos, a member of the noscapinoid family of microtubule-modulating anticancer agents.


Asunto(s)
Autofagia , Microtúbulos/metabolismo , Noscapina/análogos & derivados , Especies Reactivas de Oxígeno , Moduladores de Tubulina/metabolismo , Transporte Activo de Núcleo Celular , Antineoplásicos/farmacología , Apoptosis , Línea Celular Tumoral , Proteínas Fluorescentes Verdes/química , Humanos , Potenciales de la Membrana , Microscopía Fluorescente/métodos , Mitocondrias/metabolismo , Modelos Biológicos , Noscapina/química , Noscapina/farmacología
16.
Eur J Cancer ; 46(9): 1668-78, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20303260

RESUMEN

Hormone-refractory prostate cancer, its skeletal metastasis and complications remain a therapeutic challenge. Here we show that treatment with (S)-3-((R)-9-bromo-4-methoxy-6-methyl-5,6,7,8-tetrahydro-[1,3]dioxolo[4,5-g]isoquinolin-5-yl)-6,7-dimethoxyiso-benzofuran-1(3H)-one (EM011), the brominated analogue of a plant-derived non-toxic antitussive alkaloid, noscapine, achieved significant inhibition of hormone-refractory human prostate cancer implanted intratibially in the bone as shown by non-invasive, real-time bioluminescent imaging of tumour growth in nude mice. Mechanistically, in vitro data suggested that the antiproliferative and proapoptotic effects of EM011 in human prostate cancer cell lines were through blockade of cell-cycle progression by impairing the formation of a bipolar spindle apparatus. The G2/M arrest was accompanied by activation of the mitotic checkpoint, a pre-requisite for induction of optimal apoptosis. Attenuation of mitotic checkpoint by siRNA duplexes led to a reduction in mitotic arrest and subsequent apoptosis. Our results further demonstrated participation of an intrinsic mitochondrially mediated apoptotic pathway that ultimately triggered caspase-driven EM011-induced apoptosis. EM011 did not exert any detectable toxicity in normal tissues with frequently dividing cells such as the gut and bone marrow. Thus, these data warrant further evaluation of EM011 for the management of prostate cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Dioxoles/farmacología , Isoquinolinas/farmacología , Mitocondrias/enzimología , Neoplasias de la Próstata/tratamiento farmacológico , Moduladores de Tubulina/farmacología , Animales , Apoptosis/fisiología , Línea Celular Tumoral , Fase G2/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mitosis/efectos de los fármacos , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
17.
PLoS One ; 5(2): e9165, 2010 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-20161797

RESUMEN

Noscapine, a plant-derived, non-toxic, over-the-counter antitussive alkaloid has tubulin-binding properties. Based upon the structural resemblance of noscapine to colchicine, a tubulin-binding anti-inflammatory drug, noscapine and its semi-synthetic brominated analogs were examined for in vitro anti-inflammatory activity. Brominated noscapine analogs were found to inhibit cytokine and chemokine release from macrophage cell lines but did not affect cell viability. Brominated noscapine analogs demonstrated anti-inflammatory properties in both TLR- and non-TLR induced in vitro innate immune pathway inflammation models, mimicking septic and sterile infection respectively. In addition, electron microscopy and immunoblotting data indicated that these analogs induced robust autophagy in human macrophages. This study is the first report to identify brominated noscapines as innate immune pathway anti-inflammatory molecules.


Asunto(s)
Antiinflamatorios/farmacología , Macrófagos/efectos de los fármacos , Noscapina/farmacología , Moduladores de Tubulina/farmacología , Animales , Antiinflamatorios/química , Antitusígenos/química , Antitusígenos/farmacología , Autofagia/efectos de los fármacos , Western Blotting , Línea Celular , Quimiocina CXCL10/metabolismo , Relación Dosis-Respuesta a Droga , Halogenación , Humanos , Interleucina-8/metabolismo , Macrófagos/metabolismo , Macrófagos/ultraestructura , Microscopía Electrónica , Modelos Biológicos , Estructura Molecular , Óxido Nítrico/metabolismo , Noscapina/análogos & derivados , Noscapina/química , Especies Reactivas de Oxígeno/metabolismo , Receptor Toll-Like 4/metabolismo , Moduladores de Tubulina/química , Factor de Necrosis Tumoral alfa/metabolismo
18.
Mol Cancer ; 8: 93, 2009 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-19878573

RESUMEN

BACKGROUND: Lung cancer remains a leading cause of cancer death among both men and women in the United States. Treatment modalities available for this malignancy are inadequate and thus new drugs with improved pharmacological profiles and superior therapeutic indices are being continually explored. Noscapinoids constitute an emerging class of anticancer agents that bind tubulin but do not significantly alter the monomer/polymer ratio of tubulin. EM011, a rationally-designed member of this class of non-toxic agents, is more potent than the lead molecule, noscapine. RESULTS: Here we report that EM011 inhibited proliferation of a comprehensive panel of lung cancer cells with IC(50)'s ranging from 4-50 microM. In A549 human non-small cell lung cancer cells, the antiproliferative activity was mediated through blockage of cell-cycle progression by induction of a transient but robust mitotic arrest accompanied by activation of the spindle assembly checkpoint. The mitotically-arrested A549 cells then override the activated mitotic checkpoint and aberrantly exit mitosis without cytokinesis resulting in pseudo G1-like multinucleated cells that either succumb directly to apoptosis or continue another round of the cell-cycle. The accumulated enormous DNA perhaps acts as genotoxic stress to trigger cell death. EM011-induced apoptotic cell death in A549 cells was associated with a decrease of the Bcl2/BAX ratio, activation of caspase-3 and cleavage of PARP. Furthermore, EM011 induced downregulation of survivin expression over time of treatment. Abrogation of survivin led to an increase of cell death whereas, overexpression caused decreased apoptosis. CONCLUSION: These in vitro data suggest that EM011 mediates antiproliferative and proapoptotic activity in non-small cell A549 lung cancer cells by impeding cell-cycle progression and attenuating antiapoptotic signaling circuitries (viz. Bcl2, survivin). The study provides evidence for the potential usefulness of EM011 in chemotherapy of lung cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/patología , Dioxoles/farmacología , Isoquinolinas/farmacología , Neoplasias Pulmonares/patología , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Caspasa 3/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Fragmentación del ADN/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Activación Enzimática/efectos de los fármacos , Fase G2/efectos de los fármacos , Humanos , Proteínas Inhibidoras de la Apoptosis , Neoplasias Pulmonares/enzimología , Mitosis/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasas/metabolismo , ARN Interferente Pequeño/metabolismo , Huso Acromático/efectos de los fármacos , Huso Acromático/metabolismo , Survivin , Proteína X Asociada a bcl-2/metabolismo
19.
Clin Cancer Res ; 15(14): 4722-32, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19584158

RESUMEN

PURPOSE: Cell-surface receptor-targeted magnetic iron oxide nanoparticles provide molecular magnetic resonance imaging contrast agents for improving specificity of the detection of human cancer. EXPERIMENTAL DESIGN: The present study reports the development of a novel targeted iron oxide nanoparticle using a recombinant peptide containing the amino-terminal fragment of urokinase-type plasminogen activator (uPA) conjugated to magnetic iron oxide nanoparticles amino-terminal fragment conjugated-iron oxide (ATF-IO). This nanoparticle targets uPA receptor, which is overexpressed in breast cancer tissues. RESULTS: ATF-IO nanoparticles are able to specifically bind to and be internalized by uPA receptor-expressing tumor cells. Systemic delivery of ATF-IO nanoparticles into mice bearing s.c. and i.p. mammary tumors leads to the accumulation of the particles in tumors, generating a strong magnetic resonance imaging contrast detectable by a clinical magnetic resonance imaging scanner at a field strength of 3 tesla. Target specificity of ATF-IO nanoparticles showed by in vivo magnetic resonance imaging is further confirmed by near-IR fluorescence imaging of the mammary tumors using near-IR dye-labeled amino-terminal fragment peptides conjugated to iron oxide nanoparticles. Furthermore, mice administered ATF-IO nanoparticles exhibit lower uptake of the particles in the liver and spleen compared with those receiving nontargeted iron oxide nanoparticles. CONCLUSIONS: Our results suggest that uPA receptor-targeted ATF-IO nanoparticles have potential as molecularly targeted, dual modality imaging agents for in vivo imaging of breast cancer.


Asunto(s)
Imagen por Resonancia Magnética/métodos , Neoplasias Mamarias Experimentales/diagnóstico por imagen , Nanopartículas/administración & dosificación , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Animales , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Compuestos Férricos/química , Humanos , Aumento de la Imagen/métodos , Magnetismo , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas/química , Radiografía , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Espectroscopía Infrarroja Corta
20.
Mol Pharmacol ; 71(1): 101-11, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17035597

RESUMEN

The identification of differentially regulated apoptotic signals in normal and tumor cells allows the development of cancer cell-selective therapies. Increasing evidence shows that the inhibitor of apoptosis (IAP) proteins survivin and XIAP are highly expressed in tumor cells but are absent or have very low levels of expression in normal adult tissues. We found that inhibiting AKT activity with 10 to 100 nM deguelin, a small molecule derived from natural products, markedly reduced the levels of both survivin and XIAP, inducing apoptosis in human breast cancer cells but not in normal cells. It is noteworthy that we detected an elevated level of cleaved poly(ADP-ribose) polymerase, a signature of caspase activation, without a significant increase in caspase activity in deguelin-treated cancer cells. Our results suggest that severe down-regulation of the IAPs by deguelin releases their inhibitory activity over pre-existing active caspases present in cancer cells, inducing apoptosis without the need for further caspase activation. Because normal cells have very low levels of p-AKT, XIAP, survivin, and pre-existing caspase activity, deguelin had little effect on those cells. In addition, we found that combining deguelin with chemotherapy drugs enhanced drug-induced apoptosis selectively in human tumor cells, which suggests that deguelin has great potential for chemosensitization and could represent a new therapeutic agent for treatment of breast cancer.


Asunto(s)
Proteínas Inhibidoras de la Apoptosis/genética , Rotenona/análogos & derivados , Proteína Inhibidora de la Apoptosis Ligada a X/genética , Apoptosis , Neoplasias de la Mama/patología , División Celular , Línea Celular Tumoral , Ensayo de Unidades Formadoras de Colonias , Cartilla de ADN , Regulación hacia Abajo/efectos de los fármacos , Femenino , Genes Reporteros , Humanos , Proteínas Inhibidoras de la Apoptosis/efectos de los fármacos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Proteínas Asociadas a Microtúbulos/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Neoplasias/efectos de los fármacos , Proteínas de Neoplasias/genética , Proteínas Recombinantes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rotenona/farmacología , Survivin , Proteína Inhibidora de la Apoptosis Ligada a X/efectos de los fármacos , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...