Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Biotechnol Bioeng ; 116(9): 2393-2411, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31112285

RESUMEN

The new and rapid advancement in the complexity of biologics drug discovery has been driven by a deeper understanding of biological systems combined with innovative new therapeutic modalities, paving the way to breakthrough therapies for previously intractable diseases. These exciting times in biomedical innovation require the development of novel technologies to facilitate the sophisticated, multifaceted, high-paced workflows necessary to support modern large molecule drug discovery. A high-level aspiration is a true integration of "lab-on-a-chip" methods that vastly miniaturize cellulmical experiments could transform the speed, cost, and success of multiple workstreams in biologics development. Several microscale bioprocess technologies have been established that incrementally address these needs, yet each is inflexibly designed for a very specific process thus limiting an integrated holistic application. A more fully integrated nanoscale approach that incorporates manipulation, culture, analytics, and traceable digital record keeping of thousands of single cells in a relevant nanoenvironment would be a transformative technology capable of keeping pace with today's rapid and complex drug discovery demands. The recent advent of optical manipulation of cells using light-induced electrokinetics with micro- and nanoscale cell culture is poised to revolutionize both fundamental and applied biological research. In this review, we summarize the current state of the art for optical manipulation techniques and discuss emerging biological applications of this technology. In particular, we focus on promising prospects for drug discovery workflows, including antibody discovery, bioassay development, antibody engineering, and cell line development, which are enabled by the automation and industrialization of an integrated optoelectronic single-cell manipulation and culture platform. Continued development of such platforms will be well positioned to overcome many of the challenges currently associated with fragmented, low-throughput bioprocess workflows in biopharma and life science research.


Asunto(s)
Automatización , Productos Biológicos , Descubrimiento de Drogas , Dispositivos Laboratorio en un Chip , Humanos
2.
Mol Carcinog ; 54(8): 618-31, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24395385

RESUMEN

p66Shc functions as a longevity protein in murine and exhibits oxidase activity in regulating diverse biological activities. In this study, we investigated the role of p66Shc protein in regulating ovarian cancer (OCa) cell proliferation. Among three cell lines examined, the slowest growing OVCAR-3 cells have the lowest level of p66Shc protein. Transient transfection with p66Shc cDNA expression vector in OVCAR-3 cells increases cell proliferation. Conversely, knock-down of p66Shc by shRNA in rapidly growing SKOV-3 cells results in decreased cell growth. In estrogen (E2)-treated CaOV-3 cells, elevated p66Shc protein level correlates with ROS level, ErbB-2 and ERK/MAPK activation, and cell proliferation. Further, the E2-stimulated proliferation of CaOV-3 cells was blocked by antioxidants and ErbB-2 inhibitor. Additionally, in E2-stimulated cells, the tartrate-sensitive, but not the tartrate-resistant, phosphatase activity decreases; concurrently, the tyrosine phosphorylation of ErbB-2 increases. Conversely, inhibition of phosphatase activity by L(+)-tartrate treatment increases p66Shc protein level, ErbB-2 tyrosine phosphorylation, ERK/MAPK activation, and cell growth. Further, inhibition of the ERK/MAPK pathway by PD98059 blocks E2-induced ERK/MAPK activation and cell proliferation in CaOV-3 cells. Moreover, immunohistochemical analyses showed that the p66Shc protein level was significantly higher in cancerous cells than in noncancerous cells in archival OCa tissues (n = 76; P = 0.00037). These data collectively indicate that p66Shc protein plays a critical role in up-regulating OCa progression.


Asunto(s)
Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Receptor ErbB-2/metabolismo , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Estrógenos/farmacología , Femenino , Flavonoides/farmacología , Humanos , Neoplasias Ováricas/genética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Adaptadoras de la Señalización Shc/genética , Transducción de Señal/efectos de los fármacos , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Regulación hacia Arriba
3.
Bioorg Med Chem Lett ; 25(21): 4866-4871, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26112439

RESUMEN

Many efforts are underway to develop selective inhibitors of the voltage-gated sodium channel NaV1.7 as new analgesics. Thus far, however, in vitro selectivity has proved difficult for small molecules, and peptides generally lack appropriate pharmacokinetic properties. We previously identified the NaV1.7 inhibitory peptide GpTx-1 from tarantula venom and optimized its potency and selectivity via structure-guided analoging. To further understand GpTx-1 binding to NaV1.7, we have mapped the binding site to transmembrane segments 1-4 of the second pseudosubunit internal repeat (commonly referred to as Site 4) using NaV1.5/NaV1.7 chimeric protein constructs. We also report that select GpTx-1 amino acid residues apparently not contacting NaV1.7 can be derivatized with a hydrophilic polymer without adversely affecting peptide potency. Homodimerization of GpTx-1 with a bifunctional polyethylene glycol (PEG) linker resulted in a compound with increased potency and a significantly reduced off-rate, demonstrating the ability to modulate the function and properties of GpTx-1 by linking to additional molecules.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Péptidos/química , Péptidos/farmacología , Ingeniería de Proteínas , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Dimerización , Relación Dosis-Respuesta a Droga , Humanos , Conformación Molecular , Unión Proteica , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Bloqueadores del Canal de Sodio Activado por Voltaje/química
4.
J Pharmacol Exp Ther ; 345(2): 225-38, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23475901

RESUMEN

Calcium entry into T cells following antigen stimulation is crucial for nuclear factor of activated T cells (NFAT)-mediated T cell activation. The movement of calcium is mediated by calcium release-activated calcium (CRAC) channels. There are two key components of this channel: Orai1 is the pore-forming subunit located in the plasma membrane, and stromal interaction molecule 1 (STIM1) functions as a Ca(2+) sensor in the endoplasmic reticulum. A subset of human patients carry mutations in either STIM1 or Orai1 that affect protein function or expression, resulting in defective store-operated Ca(2+) influx and CRAC channel function, and impaired T cell activation. These patients suffer from a hereditary form of severe combined immune deficiency syndrome, highlighting the importance of the CRAC channel for T lymphocyte function in humans. Since autoreactive T cells play an important role in the development of autoimmune diseases such as rheumatoid arthritis, multiple sclerosis, and organ transplantation, Orai1 becomes an attractive therapeutic target for ameliorating autoimmune disease. We developed a novel approach to inhibiting CRAC function by generating high-affinity fully human monoclonal antibodies to human Orai1. These antibodies inhibited ICRAC current, store-operated Ca(2+) influx, NFAT transcription, and cytokine release. These fully human antibodies to human Orai1 may represent a novel therapeutic approach for the treatment of autoimmunity.


Asunto(s)
Anticuerpos Bloqueadores/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Enfermedades Autoinmunes/tratamiento farmacológico , Canales de Calcio/efectos de los fármacos , Canales de Calcio/inmunología , Aequorina/farmacología , Secuencia de Aminoácidos , Animales , Anticuerpos Bloqueadores/biosíntesis , Anticuerpos Monoclonales/biosíntesis , Western Blotting , Quimera , Citocinas/sangre , Mapeo Epitopo , Epítopos/efectos de los fármacos , Citometría de Flujo , Genes Reporteros , Células HEK293 , Humanos , Células Jurkat , Cinética , Luciferasas/genética , Ratones , Datos de Secuencia Molecular , Factores de Transcripción NFATC/biosíntesis , Factores de Transcripción NFATC/genética , Proteína ORAI1 , Técnicas de Placa-Clamp , Polimorfismo de Nucleótido Simple , Ratas
5.
J Biol Chem ; 285(31): 23598-606, 2010 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-20498373

RESUMEN

Cellular prostatic acid phosphatase (cPAcP), an authentic tyrosine phosphatase, is proposed to function as a negative growth regulator of prostate cancer (PCa) cells in part through its dephosphorylation of ErbB-2. Nevertheless, the direct interaction between cPAcP and ErbB-2 has not been shown nor the specific dephosphorylation site of ErbB-2 by cPAcP. In this report, our data show that the phosphorylation level of ErbB-2 primarily at Tyr(1221/2) correlates with the growth rate of both LNCaP and MDA PCa2b human PCa cells. Further, cPAcP reciprocally co-immunoprecipitated with ErbB-2 in a non-permissive growth condition. Expression of wild type cPAcP, but not inactive mutant, by cDNA in cPAcP-null LNCaP C-81 cells results in decreased tyrosine phosphorylation of ErbB-2 including Tyr(1221/2). Concurrently, Tyr(317) phosphorylation of p52(Shc), proliferating cell nuclear antigen expression, and cell growth are decreased in these cells. Conversely, decreased cPAcP expression by short hairpin RNA in LNCaP C-33 cells was associated with elevated phosphorylation of ErbB-2 initially at Tyr(1221/2). Its downstream p52(Shc), ERK1/2, Akt, Src, STAT-3, and STAT-5 were activated, and cell proliferation, proliferating cell nuclear antigen, and cyclin D1 expression were increased. Stable subclones of C-33 cells by small interfering PAcP had elevated Tyr(1221/2) phosphorylation of ErbB-2 and exhibited androgen-independent growth and increased tumorigenicity in xenograft female animals. In summary, our data together indicate that in prostate epithelia, cPAcP interacts with and dephosphorylates ErbB-2 primarily at Tyr(1221/2) and hence blocks downstream signaling, leading to reduced cell growth. In PCa cells, decreased cPAcP expression is associated with androgen-independent cell proliferation and tumorigenicity as seen in advanced hormone-refractory prostate carcinomas.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/metabolismo , Proteínas Tirosina Fosfatasas/fisiología , Receptor ErbB-2/metabolismo , Fosfatasa Ácida , Animales , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Masculino , Ratones , Trasplante de Neoplasias , Fosforilación , Proteínas Tirosina Fosfatasas/metabolismo , Fracciones Subcelulares , Tirosina/química
6.
J Oncol Res Ther ; 3(5)2017.
Artículo en Inglés | MEDLINE | ID: mdl-31328181

RESUMEN

The treatment of castration-resistant (CR) prostate cancer (PCa) is limited. A sub-population of CR PCa tumors can synthesize androgens for intracrine androgen receptor (AR) activation, thus targeting androgen biosynthesis could be an effective therapeutic option for these patients. We determined that androgen biosynthesis inhibitors simvastatin, atorvastatin, and ketoconazole directly inhibit growth, migration, and colony formation of LNCaP C-81 cells, which exhibit de novo androgen biosynthesis, with simvastatin being the most effective. Importantly, in combination treatments, statins specifically enhanced growth suppression with added effects by anti-androgen abiraterone acetate on the CR PCa cells. Thus, statins can be used in conjunction with abiraterone acetate to enhance anti-androgen therapy for CR PCa.

7.
Oncogene ; 24(48): 7203-12, 2005 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-16170380

RESUMEN

p66(Shc), an isoform of Shc adaptor proteins, is shown to mediate various signals, including cellular stress. However, little is known about its involvement in carcinogenesis. We previously showed that p66(Shc) protein level is upregulated by steroid hormones in human carcinoma cells and is higher in prostate cancer (PCa) specimens than adjacent noncancerous cells. In this study, we investigated the role of p66(Shc) protein in PCa cell proliferation. Among different PCa cell lines tested, p66(Shc) protein level showed positive correlation with cell proliferation, that is, rapid-growing cells expressed higher p66(Shc) protein than slow-growing cells. Exposure of slow-growing LNCaP C-33 cells to epidermal growth factor (EGF) and 5alpha-dihydrotestosterone (DHT) led to upregulation of proliferation and p66(Shc) protein level. Conversely, growth suppression of fast-growing cells by cellular form of prostatic acid phosphatase (cPAcP) expression, a negative growth regulator, down-regulated their p66(Shc) protein level. Additionally, increased expression of p66(Shc) protein by cDNA transfection in LNCaP C-33 cells resulted in increased cell proliferation. Cell cycle analyses showed higher percentage of p66(Shc)-overexpressing cells at S phase (24%) than control cells (17%), correlating with their growth rates. On the other hand, transient knock-down of p66(Shc) expression by RNAi in rapidly growing cells decreased their proliferation as evidenced by the reduced cell growth as well as S phase in p66(Shc)-knocked down cells. The p66(Shc) signaling in cell growth regulation is apparently mediated by extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK). Thus, our results indicate a novel role for p66(Shc) in prostate carcinogenesis, in part, promoting cell proliferation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proliferación Celular , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias de la Próstata/metabolismo , Fosfatasa Ácida , Proteínas Adaptadoras Transductoras de Señales/genética , Línea Celular Tumoral , Células Clonales , Dihidrotestosterona/farmacología , Factor de Crecimiento Epidérmico/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Masculino , Fosforilación , Neoplasias de la Próstata/patología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Tirosina Fosfatasas/farmacología , Interferencia de ARN , Fase S , Proteínas Adaptadoras de la Señalización Shc , Transducción de Señal , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Regulación hacia Arriba/efectos de los fármacos
8.
Endocr Relat Cancer ; 13(1): 151-67, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16601285

RESUMEN

Neuroendocrine (NE) cells are the minor cell populations in normal prostate epithelial compartments. During prostate carcinogenesis, the number of NE cells in malignant lesions increases, correlating with its tumorigenicity and hormone-refractory growth. It is thus proposed that cancerous NE cells promote prostate cancer (PCa) cell progression and its androgen-independent proliferation, although the origin of the cancerous NE cells is not clear. To investigate the role of cancerous NE cells in prostate carcinogenesis, we characterized three NE subclone cell lines-NE-1.3, NE-1.8 and NE-1.9, which were transdifferentiated from androgen-sensitive human PCa LNCaP cells by culturing in an androgen-depleted environment, resembling clinical androgen-ablation therapy. These subclone cells acquire many features of NE cells seen in clinical prostate carcinomas, for example exhibiting a neuronal morphology and expressing multiple NE markers, including neuron-specific enolase, chromogranin B, neurotensin, parathyroid hormone-related peptide, and to a lesser degree for chromogranin A, while lacking androgen receptor (AR) or prostate specific antigen (PSA) expression. These cells represent terminally differentiated stable cells because after 3 months of re-culturing in a medium containing androgenic activity, they still retained the NE phenotype and expressed NE markers. Despite these NE cells having a slow growth rate, they readily developed xenograft tumors. Furthermore, media conditioned by these NE cells exhibited a stimulatory effect on proliferation and PSA secretion by LNCaP cells in androgen-deprived conditions. Additionally, we found that receptor protein tyrosine phosphatase alpha plays a role in upregulating multiple NE markers and acquiring the NE phenotype. These NE cells thus represent cancerous NE cells and could serve as a useful cell model system for investigating the role of cancerous NE cells in hormone-refractory proliferation of PCa cells.


Asunto(s)
Adenocarcinoma/patología , Andrógenos/fisiología , Neoplasias de la Próstata/patología , Adenocarcinoma/metabolismo , Animales , Diferenciación Celular/fisiología , Cromogranina A , Cromograninas/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neurotensina/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores , Receptores Androgénicos/metabolismo , Receptores de Superficie Celular/metabolismo , Células Tumorales Cultivadas
9.
Invest Ophthalmol Vis Sci ; 47(7): 3164-77, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16799064

RESUMEN

PURPOSE: To determine whether specific dietary and synthetic flavonoids can protect human retinal pigment epithelial (RPE) cells from oxidative-stress-induced death. METHODS: The efficacy and potency were determined of a variety of dietary and synthetic flavonoids on the survival of human ARPE-19 cells and primary human RPE cells treated with either hydrogen peroxide (H2O2) or t-butyl hydroperoxide (t-BOOH). We determined the effective concentrations (EC50s) and the toxicities (LD50s) of the flavonoids after 24 hours, by using the MTT assay. The efficacy of vitamins E and C on RPE cell survival were compared under identical conditions. The ability of specific flavonoids to protect RPE cells from cell death was determined at various time intervals after the cells were exposed to oxidative stress. The ability of flavonoids to block the accumulation of intracellular reactive oxygen species was examined with dichlorofluorescein (DCF) fluorescence. Finally, the ability of flavonoids to induce phase-2 detoxifying enzymes was tested by immunoblot analysis for the transcription factor Nrf2 and the phase-2 gene product heme-oxygenase 1. RESULTS: Specific flavonoids protected human RPE cells from oxidative-stress-induced death with efficacies between 80% and 100% and potencies in the high-nanomolar and low-micromolar range. The toxicities of most of the effective flavonoids were low. The effective flavonoids included the dietary flavonoids fisetin, luteolin, quercetin, eriodictyol, baicalein, galangin and EGCG, and the synthetic flavonoids, 3,6-dihydroxy flavonol and 3,7 dihydroxy flavonol. Several flavonoids can protect RPE cells even when they are added after the cells have been exposed to oxidative stress. The flavonoids acted through an intracellular route to block the accumulation of reactive oxygen species. Many of these flavonoids induced the expression of Nrf2 and the phase-2 gene product heme-oxygenase 1 in human RPE cells. CONCLUSIONS: The results identify a select group of flavonoids that protect RPE cells from oxidative-stress-induced death with a high degree of potency and low toxicity. Many of these flavonoids also induce the expression of phase-2 detoxification proteins which could function to provide additional protection against oxidative stress. This select group of flavonoids and the foods that contain high levels of these compounds may have some clinical benefit for patients with retinal diseases associated with oxidative stress.


Asunto(s)
Citoprotección/efectos de los fármacos , Flavonoides/farmacología , Estrés Oxidativo/efectos de los fármacos , Epitelio Pigmentado Ocular/efectos de los fármacos , Western Blotting , Recuento de Células , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Dieta , Relación Dosis-Respuesta a Droga , Hemo-Oxigenasa 1/metabolismo , Humanos , Peróxido de Hidrógeno/toxicidad , Factor 2 Relacionado con NF-E2/metabolismo , Epitelio Pigmentado Ocular/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sales de Tetrazolio , Tiazoles , terc-Butilhidroperóxido/toxicidad
10.
Oncogene ; 22(5): 781-96, 2003 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-12569372

RESUMEN

The expression and secretion of prostate-specific antigen (PSA) are regulated by androgens in normal prostate secretory epithelial cells. In prostate cancer patients, the serum PSA level is usually elevated and cancer cells are initially responsive to androgens. However, those cancer cells become androgen-independent after androgen ablation therapy. In hormone-refractory cancer patients, even in an androgen-deprived environment, the circulation level of PSA rebounds and is constitutively elevated through a yet unknown mechanism. Tyrosine phosphorylation of ErbB-2 is involved in regulating the androgen-responsive phenotype of prostate cancer cells, and it is at least partly regulated by the cellular form of prostatic acid phosphatase (PAcP), a prostate-unique protein tyrosine phosphatase. We investigated the ErbB-2 signal pathway in androgen-independent PSA secretion. LNCaP C-81 cells, which are androgen-independent LNCaP cells lacking endogenous PAcP expression with a hypertyrosine phosphorylated ErbB-2, secreted a higher level of PSA in conditioned media than did androgen-sensitive LNCaP C-33 parental cells. A restored expression of cellular PAcP in C-81 cells was concurrent with a decrease in tyrophosphorylation of ErbB-2 and reduction of PSA secretion. Moreover, transient transfection of C-33 cells with the wild-type ErbB-2 or a constitutively active mutant of MEK1 cDNA resulted in an increased level of secreted PSA. The elevation of secreted PSA level by the forced expression of ErbB-2 was inhibited by an MEK inhibitor, PD98059. In C-81 cells, the expression of a dominant negative mutant of ErbB-2 reduced the secreted level of PSA. The inhibition of ErbB-2 or mitogen-activated protein (MAP) kinases by specific inhibitors AG879, AG825, or PD98059 led to a decrease in PSA secretion. Taken together, our data clearly indicate that the ErbB-2 signal pathway via MAP kinases (ERK1/2) is involved in regulating the secretion of PSA by androgen-independent human prostate cancer LNCaP C-81 cells in an androgen-depleted environment.


Asunto(s)
Quinasa 1 de Quinasa de Quinasa MAP , Antígeno Prostático Específico/metabolismo , Receptor ErbB-2/fisiología , Transducción de Señal/fisiología , Fosfatasa Ácida , Andrógenos/fisiología , División Celular/fisiología , Medios de Cultivo Condicionados/metabolismo , Humanos , Masculino , Proteínas Quinasas Activadas por Mitógenos/fisiología , Neoplasias de la Próstata , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas/fisiología , Receptor ErbB-2/antagonistas & inhibidores , Células Tumorales Cultivadas , Regulación hacia Arriba/fisiología
11.
Oncogene ; 22(43): 6704-16, 2003 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-14555984

RESUMEN

The neuroendocrine (NE) cells represent the third cell population in the normal prostate. Results of several clinical studies strongly indicate that the NE cell population is greatly increased in prostate carcinomas during androgen ablation therapy that correlates with hormone-refractory growth and poor prognosis. However, the mechanism of NE cell enrichment in prostate carcinoma remains an enigma. We investigated the molecular mechanism by which androgen-sensitive C-33 LNCaP human prostate cancer cells become NE-like cells in an androgen-reduced environment, mimicking clinical phenomenon. In the androgen-depleted condition, androgen-sensitive C-33 LNCaP cells gradually acquired the NE-like morphology and expressed an increased level of neuron-specific enolase (NSE), a classical marker of neuronal cells. Several NE-like subclone cells were established. Biochemical characterizations of these subclone cells showed that receptor-type protein-tyrosine phosphatase alpha (RPTPalpha) is elevated and ERK is constitutively activated, several folds higher than that in parental cells. In androgen-depleted condition, PD98059, an MEK inhibitor, could efficiently block not only the activation of ERK, but also the acquisition of the NE-like morphology and the elevation of NSE in C-33 LNCaP cells. In RPTPalpha cDNA-transfected C-33 LNCaP cells, ERK was activated and NSE was elevated. In those cells in the presence of PD98059, the ERK activation and NSE elevation were abolished, following a dose-response fashion. Additionally, in constitutively active MEK mutant cDNA-transfected C-33 LNCaP cells, ERK was activated and NSE level was elevated, and cells obtained the NE-like phenotype. Our data collectively indicated that RPTPalpha signaling via ERK is involved in the NE transdifferentiation of androgen-sensitive C-33 LNCaP human prostate cancer cells in the androgen-depleted condition.


Asunto(s)
Andrógenos/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Receptores de Superficie Celular , Transducción de Señal , Western Blotting , Diferenciación Celular , División Celular , ADN Complementario/metabolismo , Electroforesis en Gel de Poliacrilamida , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Proteínas Fluorescentes Verdes , Humanos , Proteínas Luminiscentes/metabolismo , Masculino , Microscopía Fluorescente , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Modelos Biológicos , Fosfopiruvato Hidratasa/biosíntesis , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores , Factores de Tiempo , Transfección , Células Tumorales Cultivadas
12.
Endocr Relat Cancer ; 12(4): 805-22, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16322323

RESUMEN

Human prostatic acid phosphatase (PAcP) was used as a valuable surrogate marker for monitoring prostate cancer prior to the availability of prostate-specific antigen (PSA). Even though the level of PAcP is increased in the circulation of prostate cancer patients, its intracellular level and activity are greatly diminished in prostate cancer cells. Recent advances in understanding the function of the cellular form of PAcP (cPAcP) have shed some light on its role in prostate carcinogenesis, which may have potential applications for prostate cancer therapy. It is now evident that cPAcP functions as a neutral protein tyrosine phosphatase (PTP) in prostate cancer cells and dephosphorylates HER-2/ErbB-2/Neu (HER-2: human epidermal growth factor receptor-2) at the phosphotyrosine (p-Tyr) residues. Dephosphorylation of HER-2 at its p-Tyr residues results in the down-regulation of its specific activity, which leads to decreases in growth and tumorigenicity of those cancer cells. Conversely, decreased cPAcP expression correlates with hyperphosphorylation of HER-2 at tyrosine residues and activation of downstream extracellular signal-regulated kinase (ERK)/mitogen activated protein kinase (MAPK) signaling, which results in prostate cancer progression as well as androgen-independent growth of prostate cancer cells. These in vitro results on the effect of cPAcP on androgen-independent growth of prostate cancer cells corroborate the clinical findings that cPAcP level is greatly decreased in advanced prostate cancer and provide insights into one of the molecular mechanisms involved in prostate cancer progression. Results from experiments using xenograft animal models further indicate a novel role of cPAcP as a tumor suppressor. Future studies are warranted to clarify the use of cPAcP as a therapeutic agent in human prostate cancer patients.


Asunto(s)
Andrógenos/metabolismo , Neoplasias de la Próstata/enzimología , Proteínas Tirosina Fosfatasas/metabolismo , Receptor ErbB-2/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Fosfatasa Ácida , Proliferación Celular , Epitelio/enzimología , Humanos , Masculino , Neoplasias de la Próstata/tratamiento farmacológico , Proteínas Tirosina Fosfatasas/química , Proteínas Tirosina Fosfatasas/uso terapéutico , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/uso terapéutico
13.
Cell Biosci ; 5: 38, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26185616

RESUMEN

BACKGROUND: Prostate cancer (PCa) is the most commonly diagnosed solid tumor and the second leading cancer death in the United States, and also one of the major cancer-related deaths in Chinese. Androgen deprivation therapy (ADT) is the first line treatment for metastatic PCa. PCa ultimately relapses with subsequent ADT treatment failure and becomes castrate-resistant (CR). It is important to develop effective therapies with a surrogate marker towards CR PCa. METHOD: Histone deacetylase (HDAC) inhibitors were examined to determine their effects in androgen receptor (AR)/cellular prostatic acid phosphatase (cPAcP)-positive PCa cells, including LNCaP C-33, C-81, C4-2 and C4-2B and MDA PCa2b androgen-sensitive and androgen-independent cells, and AR/cPAcP-negative PCa cells, including PC-3 and DU 145 cells. Cell growth was determined by cell number counting. Western blot analyses were carried out to determine AR, cPAcP and PSA protein levels. RESULTS: cPAcP protein level was increased by HDAC inhibitor treatment. Valproic acid, a HDAC inhibitor, suppressed the growth of AR/cPAcP-positive PCa cells by over 50% in steroid-reduced conditions, higher than on AR/cPAcP-negative PCa cells. Further, HDAC inhibitor pretreatments increased androgen responsiveness as demonstrated by PSA protein level quantitation. CONCLUSION: Our results clearly demonstrate that HDAC inhibitors can induce cPAcP protein level, increase androgen responsiveness, and exhibit higher inhibitory activities on AR/cPAcP-positive PCa cells than on AR/cPAcP-negative PCa cells. Upon HDAC inhibitor pretreatment, PSA level was greatly elevated by androgens. This data indicates the potential clinical importance of cPAcP serving as a useful biomarker in the identification of PCa patient sub-population suitable for HDAC inhibitor treatment.

14.
Cell Signal ; 27(11): 2261-71, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26257301

RESUMEN

While androgen deprivation therapy (ADT) reduces tumor burden, autocrine growth factor loops such as human epidermal growth factor receptor 2 (HER2/ErbB-2/neu) have been proposed to contribute to prostate cancer (PCa) survival and relapse. However, the role of ErbB-2 in regulating androgen-sensitive (AS) and castration-resistant (CR) cell proliferation remains unclear. Here, we determined the role of ErbB-2 in PCa progression and survival under steroid-reduced conditions using two independent PCa cell progression models. In AR-positive androgen-independent (AI) PCa cells that exhibit the CR phenotype, ErbB-2 was constitutively activated, compared to corresponding AS PCa cells. In AS LNCaP C-33 cells, androgen-induced ErbB-2 activation through ERK1/2 mediates PCa cell proliferation. Further, the ErbB-2-specific but not EGFR-specific inhibitor suppresses basal and androgen-stimulated cell proliferation and also blocks ERK1/2 activation. ErbB-2 ectopic expression and cPAcP siRNA transfection of LNCaP C-33 cells each increases ErbB-2 tyrosine phosphorylation, correlating with increased AI PSA secretion and cell proliferation. Conversely, trapping ErbB-2 by transfected endoplasmic reticulum-targeting ScFv5R expression vector abolished DHT-induced LNCaP C-33 cell growth. Moreover, inhibition of ErbB-2 but not EGFR in AI LNCaP C-81 and MDA PCa2b-AI PCa cells significantly abolished AI cell growth. In contrast to androgens via ErbB-2/ERK1/2 signaling in AS PCa cells, the inhibition of ErbB-2 abrogated AI cell proliferation by inhibiting the cell survival protein Akt in those AI cells. These results suggest that ErbB-2 is a prominent player in mediating the ligand-dependent and -independent activation of AR in AS and AI/CR PCa cells respectively for PCa progression and survival.


Asunto(s)
Antagonistas de Andrógenos/uso terapéutico , Receptores ErbB/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Receptor ErbB-2/metabolismo , Receptores Androgénicos/metabolismo , Fosfatasa Ácida/genética , Línea Celular Tumoral , Proliferación Celular/genética , Supervivencia Celular/genética , Retículo Endoplásmico , Receptores ErbB/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Calicreínas/metabolismo , Masculino , Fosforilación/genética , Antígeno Prostático Específico/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/genética , Transducción de Señal
15.
J Immunotoxicol ; 12(2): 164-73, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-24990272

RESUMEN

ORAI1 is the pore-forming component of calcium release-activated calcium (CRAC) channels. CRAC channels are the primary route for calcium ion (Ca(2+)) entry into T-cells following antigen stimulation. This Ca(2+) entry induces proliferation and cytokine production through activation of calcineurin and the nuclear factor of activated T-cells (NFAT) transcription factor along with subsequent cytokine-related genes. It was hypothesized that the in vivo inhibition of T-cell function by blocking ORAI1 or calcineurin would lead to similar functional consequences. To test this hypothesis the activity of 2C1.1, a fully human anti-ORAI1 monoclonal antibody, and cyclosporin A (CsA) were tested in vivo for their suppressive effect on T-cell-derived cytokine production and a T-cell-dependent antibody response (TDAR) using sheep red blood cells (SRBC) in cynomolgus monkeys. Despite showing similar inhibition of ex vivo interleukin (IL)-2 production by stimulated T-cells, both molecules exhibited different pharmacologic effects on the SRBC antibody response. CsA blocked the development of SRBC-specific antibodies, while 2C1.1 failed to inhibit the antigen-specific antibody response. These surprising observations suggest that full inhibition of the CRAC channel is required to inhibit a functional immune response, consistent with findings from human patients with loss of function mutations in ORAI1.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Canales de Calcio/metabolismo , Ciclosporina/administración & dosificación , Macaca fascicularis , Linfocitos T/inmunología , Animales , Formación de Anticuerpos , Calcineurina/metabolismo , Canales de Calcio/inmunología , Bovinos , Células Cultivadas , Eritrocitos/inmunología , Humanos , Interleucina-2/metabolismo , Activación de Linfocitos , Masculino , Factores de Transcripción NFATC/metabolismo , Proteína ORAI1 , Ovinos
16.
J Neuroimmunol ; 153(1-2): 108-21, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15265669

RESUMEN

FTY720 is a prodrug for FTY-phosphate, an agonist at four of the five known receptors for sphingosine-1-phosphate (S1P). We show that administration of either FTY720 or FTY-P to SJL mice with established relapsing-remitting experimental autoimmune encephalitis (EAE) results in a rapid and sustained improvement in their clinical status, and a reversal of changes in expression of mRNAs encoding some myelin proteins and inflammatory mediators. EAE produced by adoptively transferring lymph node cells from immunized mice to naïve hosts is similarly ameliorated by FTY-P. Treatment with FTY-P is accompanied by a dose-responsive peripheral lymphopoenia.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Inmunosupresores/uso terapéutico , Glicoles de Propileno/uso terapéutico , Receptores Acoplados a Proteínas G/agonistas , Animales , Antineoplásicos/uso terapéutico , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Encefalomielitis Autoinmune Experimental/sangre , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Clorhidrato de Fingolimod , Regulación de la Expresión Génica/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Inmunosupresores/farmacología , Interferón gamma/genética , Interferón gamma/metabolismo , Linfocitos/efectos de los fármacos , Linfocitos/fisiología , Linfopenia/tratamiento farmacológico , Linfopenia/etiología , Ratones , Ratones Endogámicos , Mitoxantrona/uso terapéutico , Datos de Secuencia Molecular , Proteínas de la Mielina/genética , Proteínas de la Mielina/metabolismo , Proteína Proteolipídica de la Mielina , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Fragmentos de Péptidos , Glicoles de Propileno/farmacología , ARN Mensajero/biosíntesis , Receptores Lisofosfolípidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Esfingosina/análogos & derivados , Factores de Tiempo
17.
Brain Res ; 990(1-2): 182-94, 2003 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-14568343

RESUMEN

Lysophosphatidic acid (1-acyl-2-lyso-sn-glycero-3-phosphate; LPA) and sphingosine-1-phosphate (S1P) are bioactive phospholipids which respectively act as agonists for the G-protein-coupled lpA receptors (LPA1, LPA2, and LPA3) and s1p receptors (S1P1, S1P2, S1P3, S1P4, and S1P5), collectively referred to as lysophospholipid receptors (lpR). Since astrocytes are responsive to LPA and S1P, we examined mechanisms of lpR signaling in rat cortical secondary astrocytes. Rat cortical astrocyte mRNA expression by quantitative TaqMan polymerase chain reaction (PCR) analysis revealed the following order of relative expression of lpR mRNAs: s1p3>s1p1>lpa1>s1p2=lpa3>>s1p5. Activation of lpRs by LPA or S1P led to multiple pharmacological effects, including the influx of calcium, phosphoinositide (PI) hydrolysis, phosphorylation of extracellular receptor regulated kinase (ERK) and release of [3H]-arachidonic acid (AA). These signalling events downstream of lpR activation were inhibited to varying degrees by pertussis toxin (PTX) pretreatment or by the inhibition of sphingosine kinase (SK), a rate-limiting enzyme in the biosynthesis of S1P from sphingosine. These results suggest that astrocyte lpR signalling mechanisms likely involve both Gi- and Gq-coupled GPCRs and that receptor-mediated activation of SK leads to intracellular generation of S1P, which in turn amplifies the lpR signalling in a paracrine/autocrine manner.


Asunto(s)
Astrocitos/fisiología , Corteza Cerebral/fisiología , Lisofosfolípidos/metabolismo , Receptores de Lipoproteína/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Esfingosina/análogos & derivados , Actinas/biosíntesis , Actinas/genética , Animales , Ácido Araquidónico/metabolismo , Astrocitos/efectos de los fármacos , Western Blotting , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Clonación Molecular , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Fosfatos de Inositol/metabolismo , Lisofosfolípidos/farmacología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Fosforilación , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Receptor Cross-Talk/efectos de los fármacos , Receptores de Lipoproteína/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Esfingosina/farmacología
18.
Int J Dev Neurosci ; 21(8): 431-43, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14659994

RESUMEN

We used a simple commercial magnetic immunobead method for the preparation of acutely isolated microglial cells from postnatal days 1-3 rat brain. With the exception of a 15 min enzyme incubation, all stages are carried out at 4 degrees C, minimizing the opportunity for changes in gene expression during the isolation to be reflected in changes in accumulated mRNA. The composition of the isolated cells was compared with that of microglial cultures prepared by conventional tissue culture methods, and the purity of microglia was comparable between the two preparations. RT-PCR analysis of several genes related to inflammatory products indicated that the acutely prepared cells were in a less activated condition than the conventionally tissue cultured cells. We examined the pattern of expression of receptors for lysophosphatidic acid (lpa) and sphingosine-1-phosphate (S1P) using quantitative real-time PCR (TaqMan PCR) techniques. mRNA for LPA1, S1P1, S1P2, S1P3 and S1P5 was detected in these preparations, but the levels of the different receptor mRNAs varied according to the state of activation of the cells. mRNA for LPA3 was only detected significantly in cultured cell after lipopolysaccharide (LPS) stimulation, being almost absent in cultured microglia and undetectable in the acutely isolated preparations. The levels of mRNA of LPA1 and S1P receptors was reduced by overnight exposure to S1P, while the same treatment significantly up-regulated the level of LPA3 mRNA.


Asunto(s)
Expresión Génica/efectos de los fármacos , Lisofosfolípidos/farmacología , Microglía/metabolismo , Receptores de Lipoproteína/biosíntesis , Animales , Animales Recién Nacidos , Astrocitos/metabolismo , Secuencia de Bases , Northern Blotting , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Células Cultivadas , Ciclooxigenasa 2 , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente/métodos , Proteína Ácida Fibrilar de la Glía/metabolismo , Imidazoles/farmacología , Indoles/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Lectinas/metabolismo , Proteína Básica de Mielina/metabolismo , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Oligodendroglía/metabolismo , Osteopontina , Prostaglandina-Endoperóxido Sintasas/genética , Prostaglandina-Endoperóxido Sintasas/metabolismo , Piridinas/farmacología , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/biosíntesis , Receptores Acoplados a Proteínas G/genética , Receptores de Lipoproteína/efectos de los fármacos , Receptores de Lipoproteína/genética , Receptores Lisofosfolípidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Sialoglicoproteínas/genética , Sialoglicoproteínas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos
19.
Int J Dev Neurosci ; 22(3): 131-5, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15140466

RESUMEN

Reactive gliosis is an aspect of neural plasticity and growth factor (GF) stimulation of astrocytes in vitro is widely regarded as a model system to study astrocyte plasticity. Astrocytes express receptors for several ligands including lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P), agonists for the G-protein-coupled lysophospholipid receptors (lpRs). Activation of lpRs by LPA or S1P leads to multiple pharmacological effects including the influx of calcium, phosphoinositide (PI) hydrolysis, phosphorylation of extracellular receptor regulated kinase (ERK), release of arachidonic acid, and induces mitogenesis. Treatment of astrocytes in vitro with a growth factor cocktail (containing epidermal growth factor [EGF], basic fibroblast growth factor [bFGF] and insulin) led to a marked attenuation of lpR-induced PI hydrolysis. In contrast, under identical conditions, GF treatment led to marked potentiation of PI hydrolysis downstream of activation of another abundantly expressed G-protein coupled receptor, mGluR5. Quantitative gene expression analysis of GF-treated or control astrocytes by TaqMan RT-PCR indicated that GF treatment did not change gene expression of lpa1 and s1p1, but increased gene expression of s1p5 which is expressed at very low levels in basal conditions. These results suggest that GF differentially affected PLC activation downstream of mGluR5 versus lpR activation and that the changes in mRNA levels of lpRs do not account for marked attenuation of agonist-induced phosphoinositide turnover.


Asunto(s)
Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Sustancias de Crecimiento/farmacología , Fosfatidilinositoles/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Factor de Crecimiento Epidérmico/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Fosfatos de Inositol/metabolismo , Insulina/farmacología , Fosforilación , Ratas , Ratas Sprague-Dawley , Receptor del Glutamato Metabotropico 5 , Receptores Lisofosfolípidos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
20.
J Histochem Cytochem ; 62(12): 864-78, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25249026

RESUMEN

We determined the expression of ORAI1 protein in rodent and non-rodent tissues using a monoclonal antibody directed against an extracellular loop of the protein. Previous reports using antibodies directed at the C-terminus of ORAI1 have not detected central nervous system (CNS) expression. Our results demonstrate broad tissue expression that includes the CNS using a unique monoclonal antibody specific to an extracellular loop of ORAI1. In addition, we present in situ hybridization (ISH) results using a probe within the middle of the mouse coding region showing CNS expression of Orai1 RNA. We contrast the patterns of rodent and human tissue expression and conclude that rodents have similar expression of ORAI1 in most tissue types when compared to primates, with an important exception being the male reproductive system, where human-specific expression is observed.


Asunto(s)
Canales de Calcio/análisis , Inmunohistoquímica/métodos , Animales , Anticuerpos Monoclonales/análisis , Canales de Calcio/genética , Línea Celular , Sistema Nervioso Central/química , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/ultraestructura , Femenino , Humanos , Hibridación in Situ/métodos , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína ORAI1 , ARN Mensajero/análisis , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Análisis de Matrices Tisulares/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA