Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 168
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 117(32): 19435-19445, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32719131

RESUMEN

The Ras/RAF/MEK/ERK pathway is an essential signaling cascade for various refractory cancers, such as those with mutant KRAS (mKRAS) and BRAF (mBRAF). However, there are unsolved ambiguities underlying mechanisms for this growth signaling thereby creating therapeutic complications. This study shows that a vital component of the pathway CRAF is directly impacted by an end product of the cascade, glutathione transferases (GST) P1 (GSTP1), driving a previously unrecognized autocrine cycle that sustains proliferation of mKRAS and mBRAF cancer cells, independent of oncogenic stimuli. The CRAF interaction with GSTP1 occurs at its N-terminal regulatory domain, CR1 motif, resulting in its stabilization, enhanced dimerization, and augmented catalytic activity. Consistent with the autocrine cycle scheme, silencing GSTP1 brought about significant suppression of proliferation of mKRAS and mBRAF cells in vitro and suppressed tumorigenesis of the xenografted mKRAS tumor in vivo. GSTP1 knockout mice showed significantly impaired carcinogenesis of mKRAS colon cancer. Consequently, hindering the autocrine loop by targeting CRAF/GSTP1 interactions should provide innovative therapeutic modalities for these cancers.


Asunto(s)
Gutatión-S-Transferasa pi/metabolismo , Neoplasias/patología , Proteínas Proto-Oncogénicas c-raf/metabolismo , Animales , Carcinogénesis , Línea Celular Tumoral , Proliferación Celular , Gutatión-S-Transferasa pi/antagonistas & inhibidores , Gutatión-S-Transferasa pi/deficiencia , Gutatión-S-Transferasa pi/genética , Humanos , Ratones , Ratones Noqueados , Mutación , Neoplasias/genética , Neoplasias/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas/genética , Multimerización de Proteína , Estabilidad Proteica , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas c-raf/química , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Proteínas Proto-Oncogénicas p21(ras)/genética , Transducción de Señal
2.
Artículo en Inglés | MEDLINE | ID: mdl-35153270

RESUMEN

Glutathione-S transferase P1 (GSTP1) is one of the glutathione-S transferase isozymes that belong to a family of phase II metabolic isozymes. The unique feature of GSTP1 compared with other GST isozymes is its relatively high expression in malignant tissues. Thus, clinically, GSTP1 serves as a tumor marker and as a refractory factor against certain types of anticancer drugs through its primary function as a detoxifying enzyme. Additionally, recent studies have identified a chaperone activity of GSTP1 involved in the regulation the function of various intracellular proteins, including factors of the growth signaling pathway. In this review, we will first describe the function of GSTP1 and then extend the details onto its role in the mitogen-activated protein kinase signal pathway, referring to the results of our recent study that proposed a novel autocrine signal loop formed by the CRAF/GSTP1 complex in mutated KRAS and BRAF cancers. Finally, the possibilities of new therapeutic approaches for these cancers by targeting this complex will be discussed.


Asunto(s)
Antineoplásicos , Gutatión-S-Transferasa pi , Línea Celular Tumoral , Glutatión , Gutatión-S-Transferasa pi/metabolismo , Glutatión Transferasa
3.
Invest New Drugs ; 39(6): 1484-1492, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34031785

RESUMEN

Pancreatic cancer is among the most refractory malignancies with poor prognosis. Thus, preventive approaches, in addition to the development of novel therapeutic strategies are essential for this type of cancer. KRAS mutations occur very early in the development of pancreatic cancers and could be targeted for its prevention, yet specific inhibitors for mutated KRAS are lacking. Accordingly, Glutathione-S Transferase p1 (GSTP1), which we recently found to be an autocrine stimulator of mutated KRAS signaling, is predicted to be an alternative target for chemoprevention of pancreatic cancer. In this study, chemopreventive effects of O-Hexadecyl-γ-glutamyl-S-benzyl-cysteinyl-D-phenyl glycine-Ethylester (HGBPE), which we previously synthesized to inhibit GSTP1 activity, was analyzed for its effect on the prevention of a rat pancreatic carcinogenesis model induced by 7,12-dimethyl-benzanthracene (DMBA). Rats administered with DMBA were grouped into five cohorts. In the treated group I, which was treated neither with HGBPE nor vehicle, sequential appearance of precancerous lesions, ductal complexes, and adenocarcinoma was confirmed as previously reported. We also confirmed in this group that mutations of KRAS and expression of GSTP1 simultaneously occurred in the ductal complex. To rats of groups II and IV, HGBPE was administered, and vehicle to those of group III and V. In groups of II and IV, the incidence of both ductal complex and adenocarcinoma were significantly lower than those in groups III and V. These data clearly suggest the efficacy of HGBP as a potential chemopreventive agent for pancreatic cancer.


Asunto(s)
Gutatión-S-Transferasa pi/farmacología , Neoplasias Pancreáticas/prevención & control , Proteínas Proto-Oncogénicas p21(ras)/efectos de los fármacos , 9,10-Dimetil-1,2-benzantraceno/farmacología , Animales , Modelos Animales de Enfermedad , Neoplasias Pancreáticas/inducido químicamente , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
4.
J Gastroenterol Hepatol ; 36(12): 3418-3428, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34151462

RESUMEN

BACKGROUND AND AIM: In chronic hepatic diseases where treatment strategies are not available, deposited fibrotic tissues deteriorate the intrinsic regeneration capacity of the liver by creating special restrictions. Thus, if the anti-fibrosis modality is efficiently applied, the regeneration capacity of the liver should be reactivated even in such refractory hepatic diseases. METHODS: Rat liver fibrosis was induced by dimethyl-nitrosamine (DMN). Another liver fibrosis model was established in CCl4 treated Sox9CreERT2ROSA26: YFP mice. To resolve hepatic fibrosis, vitamin A-coupled liposomes containing siRNA HSP47 (VA-liposome siHSP47) were employed. EpCAM + hepatic progenitor cells from GFP rats were transplanted to DMN rat liver to examine their trans-differentiation into hepatic cells after resolution of liver fibrosis. RESULTS: Even under continuous exposure to such strong hepatotoxin as DMN, rats undergoing VA-liposome siHSP47 treatment showed an increment of DNA synthesis of hepatocytes with the concomitant restoration of impaired liver weight and normalization of albumin levels. These results were consistent with the observation that GFP + EpCAM hepatic progenitor cells transplanted to DMN rat liver, trans-differentiated into GFP + mature hepatic cells after VA-liposome siHSP47 treatment. Another rodent model also proved regeneration potential of the fibrotic liver in CCl4 administered Sox9CreERT2ROSA26: YFP mice, VA-liposome siHSP47 treatment-induced restoration of liver weight and trans-differentiation of YEP + Sox9 + cells into YFP + hepatic cells, although because of relatively mild hepatotoxicity of CCl4, undamaged hepatocytes also proliferated. CONCLUSIONS: These results demonstrated that regeneration of chronically damaged liver indeed occurs after anti-fibrosis treatment even under continuous exposure to hepatotoxin, which promises a significant benefit of the anti-fibrosis therapy for refractory liver diseases.


Asunto(s)
Liposomas , Cirrosis Hepática , ARN Interferente Pequeño , Vitamina A , Animales , Fibrosis , Liposomas/farmacología , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/patología , Regeneración Hepática/efectos de los fármacos , Lesión Pulmonar/patología , Ratones , ARN Interferente Pequeño/farmacología , Ratas , Resultado del Tratamiento , Vitamina A/farmacología
5.
Blood ; 131(13): 1476-1485, 2018 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-29363541

RESUMEN

Chronic graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell transplantation (SCT) is characterized by multiorgan fibrosis and profoundly affects the quality of life of transplant survivors. Heat shock protein 47 (HSP47), a collagen-specific molecular chaperone, plays a critical role in collagen synthesis in myofibroblasts. We explored the role of HSP47 in the fibrotic process of cutaneous chronic GVHD in mice. Immunohistochemical analysis showed massive fibrosis with elevated amounts of collagen deposits and accumulation of F4/80+ macrophages, as well as myofibroblasts expressing HSP47 and retinol-binding protein 1 in the skin after allogeneic SCT. Repeated injection of anti-colony-stimulating factor (CSF-1) receptor-blocking antibodies significantly reduced HSP47+ myofibroblasts in the skin, indicating a macrophage-dependent accumulation of myofibroblasts. Vitamin A-coupled liposomes carrying HSP47 small interfering RNA (siRNA) (VA-lip HSP47) delivered HSP47 siRNA to cells expressing vitamin A receptors and knocked down their HSP47 in vitro. Intravenously injected VA-lip HSP47 were specifically distributed to skin fibrotic lesions and did not affect collagen synthesis in healthy skin. VA-lip HSP47 knocked down HSP47 expression in myofibroblasts and significantly reduced collagen deposition without inducing systemic immunosuppression. It also abrogated fibrosis in the salivary glands. These results highlight a cascade of fibrosis in chronic GVHD; macrophage production of transforming growth factor ß mediates fibroblast differentiation to HSP47+ myofibroblasts that produce collagen. VA-lip HSP47 represent a novel strategy to modulate fibrosis in chronic GVHD by targeting HSP47+ myofibroblasts without inducing immunosuppression.


Asunto(s)
Enfermedad Injerto contra Huésped , Proteínas del Choque Térmico HSP47/antagonistas & inhibidores , ARN Interferente Pequeño/farmacología , Enfermedades de la Piel , Vitamina A/farmacología , Aloinjertos , Animales , Enfermedad Crónica , Colágeno , Femenino , Fibrosis , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/patología , Proteínas del Choque Térmico HSP47/genética , Proteínas del Choque Térmico HSP47/metabolismo , Trasplante de Células Madre Hematopoyéticas , Liposomas , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Miofibroblastos/metabolismo , Miofibroblastos/patología , ARN Interferente Pequeño/genética , Enfermedades de la Piel/tratamiento farmacológico , Enfermedades de la Piel/genética , Enfermedades de la Piel/metabolismo , Enfermedades de la Piel/patología
6.
Exp Lung Res ; 43(6-7): 271-282, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29035148

RESUMEN

BACKGROUND: Pulmonary fibrosis is a life-threatening pathological state of progressive interstitial lung diseases, such as idiopathic pulmonary fibrosis. Myofibroblasts are known to play a critical role in the pathogenesis of pulmonary fibrosis. This study aimed to evaluate the inhibitory effect of a small interfering RNA (siRNA) on a collagen-specific chaperone heat shock protein 47 (HSP47). The siRNA was preferentially delivered to myofibroblasts in a bleomycin (BLM)-induced pulmonary fibrosis rat model using siRNA against HSP47, encapsulated in a vitamin A-coupled liposome (VA-lip-siRNA HSP47). METHODS AND RESULTS: Male Sprague-Dawley rats were treated with an intratracheal injection of BLM or phosphate buffered saline followed by an intravenous injection of VA-lip-siRNA HSP47 three times per week under preventive administration schedules from day 1 to day 21 and therapeutic administration schedules from day 15 to day 35. The expression of HSP47 after the treatment was assessed by immunoblotting. The specific delivery of VA-lip-siRNA HSP47 conjugated with 6'-carboxyfluoresce into myofibroblasts was examined by immunofluorescence staining. The effect of VA-lip-siRNA HSP47 on fibrosis was analyzed by morphological and biochemical methods. Preferential delivery of VA-lip-siRNA HSP47 to myofibroblasts in fibrotic areas in BLM-treated rats was verified by immunofluorescence staining. Treatment of VA-lip-siRNA HSP47 clearly suppressed HSP47 expression and induced apoptosis of myofibroblasts in the lung of BLM-treated rats. Hydroxyproline levels and inflammatory cytokines in the lungs, and the number of inflammatory cells in the bronchial alveolar lavage of BLM-treated rats were significantly suppressed by the treatment. Morphological assessment showed that VA-lip-siRNA HSP47 also significantly improved the morphological pulmonary fibrosis of BLM-treated rats in both preventive and therapeutic schedules. CONCLUSIONS: These results suggest that VA-lip-siRNA HSP47 improves pulmonary fibrosis in not only preventive, but also therapeutic schedules, and thus, this drug delivery system should provide a novel therapy for refractory pulmonary fibrosis.


Asunto(s)
Colágeno/metabolismo , Proteínas del Choque Térmico HSP47/metabolismo , Liposomas/farmacología , Chaperonas Moleculares/metabolismo , Fibrosis Pulmonar/tratamiento farmacológico , ARN Interferente Pequeño/farmacología , Vitamina A/farmacología , Animales , Bleomicina/farmacología , Líquido del Lavado Bronquioalveolar/química , Citocinas/metabolismo , Hidroxiprolina/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Masculino , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Fibrosis Pulmonar/metabolismo , Ratas , Ratas Sprague-Dawley
7.
Exp Cell Res ; 341(1): 8-17, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26812497

RESUMEN

Transdifferentiation of vitamin A-storing hepatic stellate cells (HSCs) to vitamin A-depleted myofibroblastic cells leads to liver fibrosis. Vitamin A regulates lipid accumulation and gene transcription, suggesting that vitamin A is involved in the maintenance of HSC quiescence under a physiological condition. However, the precise mechanism remains elusive because there is no appropriate in vitro culture system for quiescent HSCs. Here, we show that treatment of quiescent HSCs with vitamin A partially maintained the accumulation of lipid droplets and expression of quiescent HSC markers (glial fibrillary acidic protein, peroxisome proliferator-activator receptor-γ and CCAAT/enhancer-binding protein-α) and also the expression of myofibroblastic markers (α-smooth muscle actin, heat shock protein 47 and collagen type I). On the other hand, combined treatment with vitamin A and insulin sustained the characteristic of HSC quiescence and completely suppressed the expression of myofibroblastic markers through activation of the JAK2/STAT5 signaling pathway and increased expression of sterol regulatory element binding protein-1. These treated HSCs transdifferentiated to myofibroblastic cells under a culture condition with fetal bovine serum. The results suggest an important role of vitamin A and insulin in the maintenance of HSC quiescence under a physiological condition.


Asunto(s)
Células Estrelladas Hepáticas/citología , Células Estrelladas Hepáticas/efectos de los fármacos , Insulina/farmacología , Vitamina A/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Masculino , Ratas , Ratas Sprague-Dawley
8.
J Biol Chem ; 289(29): 20209-21, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24867951

RESUMEN

Stellate cells are distributed throughout organs, where, upon chronic damage, they become activated and proliferate to secrete collagen, which results in organ fibrosis. An intriguing property of hepatic stellate cells (HSCs) is that they undergo apoptosis when collagen is resolved by stopping tissue damage or by treatment, even though the mechanisms are unknown. Here we disclose the fact that HSCs, normal diploid cells, acquired dependence on collagen for their growth during the transition from quiescent to active states. The intramolecular RGD motifs of collagen were exposed by cleavage with their own membrane type 1 matrix metalloproteinase (MT1-MMP). The following evidence supports this conclusion. When rat activated HSCs (aHSCs) were transduced with siRNA against the collagen-specific chaperone gp46 to inhibit collagen secretion, the cells underwent autophagy followed by apoptosis. Concomitantly, the growth of aHSCs was suppressed, whereas that of quiescent HSCs was not. These in vitro results are compatible with the in vivo observation that apoptosis of aHSCs was induced in cirrhotic livers of rats treated with siRNAgp46. siRNA against MT1-MMP and addition of tissue inhibitor of metalloproteinase 2 (TIMP-2), which mainly inhibits MT1-MMP, also significantly suppressed the growth of aHSCs in vitro. The RGD inhibitors echistatin and GRGDS peptide and siRNA against the RGD receptor αVß1 resulted in the inhibition of aHSCs growth. Transduction of siRNAs against gp46, αVß1, and MT1-MMP to aHSCs inhibited the survival signal of PI3K/AKT/IκB. These results could provide novel antifibrosis strategies.


Asunto(s)
Colágeno/metabolismo , Células Estrelladas Hepáticas/citología , Células Estrelladas Hepáticas/metabolismo , Metaloproteinasa 14 de la Matriz/metabolismo , Animales , Apoptosis , Proliferación Celular , Supervivencia Celular , Colágeno/antagonistas & inhibidores , Colágeno/química , Proteínas del Choque Térmico HSP47/antagonistas & inhibidores , Proteínas del Choque Térmico HSP47/genética , Proteínas del Choque Térmico HSP47/metabolismo , Células Estrelladas Hepáticas/efectos de los fármacos , Humanos , Proteínas I-kappa B/metabolismo , Integrinas/antagonistas & inhibidores , Integrinas/genética , Integrinas/metabolismo , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Ratones , Oligopéptidos/química , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Inhibidor Tisular de Metaloproteinasa-1/antagonistas & inhibidores , Inhibidor Tisular de Metaloproteinasa-1/genética , Inhibidor Tisular de Metaloproteinasa-1/metabolismo
9.
Gut ; 62(9): 1328-39, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23172890

RESUMEN

BACKGROUND AND OBJECTIVE: Fibrosis associated with chronic pancreatitis is an irreversible lesion that can disrupt pancreatic exocrine and endocrine function. Currently, there are no approved treatments for this disease. We previously showed that siRNA against collagen-specific chaperone protein gp46, encapsulated in vitamin A-coupled liposomes (VA-lip-siRNAgp46), resolved fibrosis in a model of liver cirrhosis. This treatment was investigated for pancreatic fibrosis induced by dibutyltin dichloride (DBTC) and cerulein in rats. METHODS: Specific uptake of VA-lip-siRNAgp46, conjugated with 6'-carboxyfluorescein (FAM) by activated pancreatic stellate cells (aPSCs), was analysed by fluorescence activated cell sorting (FACS). Intracellular distribution of VA-lip-siRNAgp46-FAM was examined by fluorescent microscopy. Suppression of gp46 expression by VA-lip-siRNAgp46 was assessed by immunoblotting. Collagen synthesis in aPSCs was assayed by dye-binding. Specific delivery of VA-lip-siRNAgp46 to aPSCs in DBTC rats was verified following intravenous VA-lip-siRNA-FAM and (3)H-VA-lip-siRNAgp46. The effect of VA-lip-siRNA on pancreatic histology in DBTC- and cerulein-treated rats was determined by Azan-Mallory staining and hydroxyproline content. RESULTS: FACS analysis revealed specific uptake of VA-lip-siRNAgp46-FAM through the retinol binding protein receptor by aPSCs in vitro. Immunoblotting and collagen assay verified knockdown of gp46 and suppression of collagen secretion, respectively, by aPSCs after transduction of VA-lip-siRNAgp46. Specific delivery of VA-lip-siRNAgp46 to aPSCs in fibrotic areas in DBTC rats was confirmed by fluorescence and radioactivity 24 h after the final injection. 10 systemic VA-lip-siRNAgp46 treatments resolved pancreatic fibrosis, and suppressed tissue hydroxyproline levels in DBTC- and cerulein-treated rats. CONCLUSION: These data suggest the therapeutic potential of the present approach for reversing pancreatic fibrosis.


Asunto(s)
Colágeno/biosíntesis , Fibrosis/tratamiento farmacológico , Proteínas del Choque Térmico HSP47/antagonistas & inhibidores , Páncreas/patología , Pancreatitis Crónica/complicaciones , ARN Interferente Pequeño/administración & dosificación , Animales , Ceruletida/farmacología , Fibrosis/etiología , Fármacos Gastrointestinales/farmacología , Humanos , Inmunosupresores/farmacología , Liposomas , Masculino , Modelos Animales , Compuestos Orgánicos de Estaño/farmacología , Ratas , Ratas Endogámicas Lew , Ratas Wistar , Resultado del Tratamiento , Vitamina A/farmacología , Vitaminas/farmacología
10.
J Cell Biol ; 176(7): 1049-60, 2007 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-17389234

RESUMEN

Under various pathological conditions, including infection, malignancy, and autoimmune diseases, tissues are incessantly exposed to reactive oxygen species produced by infiltrating inflammatory cells. We show augmentation of motility associated with morphological changes of human squamous carcinoma SASH1 cells, human peripheral monocytes (hPMs), and murine macrophage-like cell line J774.1 by superoxide stimulation. We also disclose that motility of hPMs and J774.1 induced by a chemotactic peptide (N-formyl-methionyl-leucyl-phenylalanine [fMLP]) was inhibited by superoxide dismutase or N-acetylcystein, indicating stimulation of motility by superoxide generated by fMLP stimulation. In these cells, protein kinase C (PKC) zeta was activated to phosphorylate RhoGDI-1, which liberated RhoGTPases, leading to their activation. These events were inhibited by dominant-negative PKCzeta in SASH1 cells, myristoylated PKCzeta peptides in hPMs and J774.1, or a specific inhibitor of RhoGTPase in SASH1, hPMs, and J774.1. These results suggest a new approach for manipulation of inflammation as well as tumor cell invasion by targeting this novel signaling pathway.


Asunto(s)
Movimiento Celular/fisiología , N-Formilmetionina Leucil-Fenilalanina/análogos & derivados , Proteína Quinasa C/metabolismo , Transducción de Señal/fisiología , Superóxidos/farmacología , Acetilcisteína/farmacología , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Inhibidores de Disociación de Guanina Nucleótido/metabolismo , Humanos , Inflamación/metabolismo , Ratones , N-Formilmetionina Leucil-Fenilalanina/farmacología , Invasividad Neoplásica , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/genética , Transducción de Señal/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa/farmacología , Superóxidos/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Inhibidor alfa de Disociación del Nucleótido Guanina rho , Inhibidores de la Disociación del Nucleótido Guanina rho-Específico
11.
Ann Hematol ; 91(3): 449-57, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21894476

RESUMEN

This prospective multicenter study was performed to clarify the efficacy and safety of micafungin (MCFG) as an empirical antifungal therapy for suspected fungal infection in patients with hematological disorders and neutropenia. Three hundred and eighty-eight patients were enrolled; 151 patients with possible fungal infection diagnosed by radiological imaging or serological testing and 237 patients with refractory fever were included in this study. The mean dose and duration of treatment with MCFG were 154.6 mg/day and 14.0 days, respectively. The clinical response rate for patients with possible fungal infection and refractory fever was 60.1% and 65.3%, respectively. Even in persistent neutropenic patients with a neutrophil count of <500/µL throughout the MCFG treatment, the clinical response rate was 46.9%. Ninety-one drug-related adverse events (DAEs) were observed in 56 patients (14.4%) and 9 serious DAEs were observed in 6 patients (1.5%). Neither daily dose nor duration of MCFG treatment affected the incidence of DAEs. It was confirmed that MCFG has adequate clinical efficacy and is safe for the treatment of suspected fungal infections in patients with hematological disorders and neutropenia.


Asunto(s)
Antifúngicos/uso terapéutico , Equinocandinas/uso terapéutico , Enfermedades Hematológicas/complicaciones , Lipopéptidos/uso terapéutico , Micosis/tratamiento farmacológico , Micosis/etiología , Neutropenia/complicaciones , Resultado del Tratamiento , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Enfermedades Hematológicas/fisiopatología , Humanos , Japón , Masculino , Micafungina , Persona de Mediana Edad , Neutropenia/fisiopatología , Estudios Prospectivos , Adulto Joven
12.
Brain ; 134(Pt 6): 1790-807, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21493695

RESUMEN

Transplantation of human mesenchymal stem cells has been shown to reduce infarct size and improve functional outcome in animal models of stroke. Here, we report a study designed to assess feasibility and safety of transplantation of autologous human mesenchymal stem cells expanded in autologous human serum in stroke patients. We report an unblinded study on 12 patients with ischaemic grey matter, white matter and mixed lesions, in contrast to a prior study on autologous mesenchymal stem cells expanded in foetal calf serum that focused on grey matter lesions. Cells cultured in human serum expanded more rapidly than in foetal calf serum, reducing cell preparation time and risk of transmissible disorders such as bovine spongiform encephalomyelitis. Autologous mesenchymal stem cells were delivered intravenously 36-133 days post-stroke. All patients had magnetic resonance angiography to identify vascular lesions, and magnetic resonance imaging prior to cell infusion and at intervals up to 1 year after. Magnetic resonance perfusion-imaging and 3D-tractography were carried out in some patients. Neurological status was scored using the National Institutes of Health Stroke Scale and modified Rankin scores. We did not observe any central nervous system tumours, abnormal cell growths or neurological deterioration, and there was no evidence for venous thromboembolism, systemic malignancy or systemic infection in any of the patients following stem cell infusion. The median daily rate of National Institutes of Health Stroke Scale change was 0.36 during the first week post-infusion, compared with a median daily rate of change of 0.04 from the first day of testing to immediately before infusion. Daily rates of change in National Institutes of Health Stroke Scale scores during longer post-infusion intervals that more closely matched the interval between initial scoring and cell infusion also showed an increase following cell infusion. Mean lesion volume as assessed by magnetic resonance imaging was reduced by >20% at 1 week post-cell infusion. While we would emphasize that the current study was unblinded, did not assess overall function or relative functional importance of different types of deficits, and does not exclude placebo effects or a contribution of recovery as a result of the natural history of stroke, our observations provide evidence supporting the feasibility and safety of delivery of a relatively large dose of autologous mesenchymal human stem cells, cultured in autologous human serum, into human subjects with stroke and support the need for additional blinded, placebo-controlled studies on autologous mesenchymal human stem cell infusion in stroke.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Madre Mesenquimatosas/fisiología , Accidente Cerebrovascular/cirugía , Adulto , Anciano , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Femenino , Estudios de Seguimiento , Humanos , Infusiones Intravenosas/métodos , Angiografía por Resonancia Magnética , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Examen Neurológico , Radiografía , Índice de Severidad de la Enfermedad , Accidente Cerebrovascular/diagnóstico por imagen , Accidente Cerebrovascular/patología , Factores de Tiempo , Trasplante Autólogo
13.
Nat Med ; 9(9): 1158-65, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12897778

RESUMEN

Bone-marrow minimal residual disease (MRD) causes relapse after chemotherapy in patients with acute myelogenous leukemia (AML). We postulate that the drug resistance is induced by the attachment of very late antigen (VLA)-4 on leukemic cells to fibronectin on bone-marrow stromal cells. We found that VLA-4-positive cells acquired resistance to anoikis (loss of anchorage) or drug-induced apoptosis through the phosphatidylinositol-3-kinase (PI-3K)/AKT/Bcl-2 signaling pathway, which is activated by the interaction of VLA-4 and fibronectin. This resistance was negated by VLA-4-specific antibodies. In a mouse model of MRD, we achieved a 100% survival rate by combining VLA-4-specific antibodies and cytosine arabinoside (AraC), whereas AraC alone prolonged survival only slightly. In addition, overall survival at 5 years was 100% for 10 VLA-4-negative patients and 44.4% for 15 VLA-4-positive patients. Thus, the interaction between VLA-4 on leukemic cells and fibronectin on stromal cells may be crucial in bone marrow MRD and AML prognosis.


Asunto(s)
Fibronectinas/metabolismo , Integrina alfa4beta1/metabolismo , Leucemia Mieloide Aguda/metabolismo , Proteínas Serina-Treonina Quinasas , Animales , Anticuerpos/farmacología , Antimetabolitos Antineoplásicos/uso terapéutico , Citarabina/uso terapéutico , Resistencia a Antineoplásicos , Humanos , Integrina alfa4beta1/efectos de los fármacos , Integrina alfa4beta1/inmunología , Integrina alfa5beta1/metabolismo , Leucemia/tratamiento farmacológico , Leucemia/metabolismo , Leucemia/patología , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/mortalidad , Ratones , Ratones SCID , Neoplasia Residual , Fosfatidilinositol 3-Quinasas/metabolismo , Valor Predictivo de las Pruebas , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal , Células del Estroma/metabolismo , Tasa de Supervivencia , Factores de Tiempo , Células Tumorales Cultivadas
14.
Cancer Sci ; 100(5): 948-55, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19245435

RESUMEN

Aberrant reactivation of hedgehog (Hh) signaling has been described in a wide variety of human cancers and in cancer stem cells. However, the contribution of Hh signaling to leukemic cell regulation has remained unclear. In this study, we assessed the possibility that Hh pathway activation contributes to the survival and drug resistance of cluster of differentiation (CD)34+ leukemia cells. Hh signaling in leukemic cell lines and primary leukemic cells was screened by reverse transcription - polymerase chain reaction (RT-PCR) and a Hh signaling reporter assay. We found that Hh signaling is active in several human acute myeloid leukemia (AML) cells, especially primary CD34+ leukemic cells and cytokine-responsive CD34+ cell lines such as Kasumi-1, Kasumi-3 and TF-1. These CD34+ cells express the downstream effectors glioma-associated oncogene homolog (GLI)1 or GLI2, indicative of active Hh signaling. Moreover, inhibition of Hh signaling with the naturally derived Smoothened antagonist cyclopamine, endogenous Hh inhibitor hedgehog-interacting protein or anti-hedgehog neutralizing antibody induced apoptosis after 48 h of exposure, although these CD34+ cell lines exhibited resistance to cytarabine (Ara-C). In contrast, cyclopamine failed to affect growth or survival in U937 and HL-60 cell lines that lack expression of Hh receptor components, confirming that the effect of Hh inhibition is specific. Furthermore, combination with 10 microM cyclopamine significantly reduced drug resistance of CD34+ cell lines and primary CD34+ leukemic cells to Ara-C. These results suggest that aberrant Hh pathway activation is a feature of some CD34+ myeloid leukemic cells and Hh inhibitors may have a therapeutic role in the treatment of AML.


Asunto(s)
Antígenos CD34/inmunología , Antígenos CD34/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Proteínas Hedgehog/antagonistas & inhibidores , Leucemia/metabolismo , Leucemia/patología , Anticuerpos/inmunología , Anticuerpos/farmacología , Médula Ósea/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas Hedgehog/metabolismo , Humanos , Leucemia/inmunología , ARN Mensajero/genética , Receptores de Superficie Celular/metabolismo , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas , Alcaloides de Veratrum/farmacología
15.
Hepatology ; 48(3): 828-40, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18756484

RESUMEN

UNLABELLED: Although conditionally replicable adenovirus (CRAd) has been used in the clinical treatment of hepatocellular carcinoma (HCC), it suffers from the inherent drawback of having relatively low antitumor activity. Here, we have sought to overcome this drawback. First, we combined CRAd (AdAFPep/Rep) driven by alpha-fetoprotein enhancer/promoter (AFPep) with a replication-incompetent adenovirus carrying a p53 transgene that is also driven by AFPep. The synergism of this combination produced a significantly improved tumoricidal effect on the human HCC cell line Hep3B, which has a relatively short doubling time in comparison with other human HCC cell lines, through the transactivation of p53 by early region 1A transcribed by AdAFPep/Rep. This synergistic interaction was augmented by the addition of a subtumoricidal dose (0.5 microg/mL) of 5-fluorouracil (5-FU), which enhanced p53 expression and facilitated the release of virions from tumor cells. When relatively large (10-mm-diameter) Hep3B tumors grown in nude mice were injected with the two viruses in combination, they showed significantly impaired growth in comparison with those treated with each virus separately. The growth suppression effect of the virus combination was enhanced by a low dose (600 microg) of 5-FU. Survival of the tumor-bearing mice treated with these three agents was significantly longer than that of control mice. Moreover, the tumor completely disappeared with the repeated injection of these agents. CONCLUSION: This combination strategy holds promise for the treatment of relatively large and rapidly growing HCCs that may be encountered clinically.


Asunto(s)
Adenoviridae/genética , Carcinoma Hepatocelular/terapia , Fluorouracilo/uso terapéutico , Neoplasias Hepáticas/terapia , Virus Oncolíticos/genética , Proteína p53 Supresora de Tumor/genética , alfa-Fetoproteínas/genética , Animales , Antimetabolitos Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/virología , Línea Celular Tumoral , Células Cultivadas , Terapia Combinada , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/virología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Viroterapia Oncolítica/métodos , Resultado del Tratamiento , Proteína p53 Supresora de Tumor/metabolismo , Replicación Viral/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto , alfa-Fetoproteínas/metabolismo
16.
Stem Cells ; 26(2): 534-42, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17962696

RESUMEN

Indian hedgehog (Ihh) plays an essential role in angiogenesis, hematogenesis, and epiphysis formation during embryogenesis. In the present study, we injected an adenoviral vector (Adv) carrying the mock-control (Adv-control) or Ihh (Adv-Ihh) gene into severe combined immunodeficiency (SCID) or BALB/c mice to evaluate the effects of lhh on the regulation of postnatal hematopoiesis in vivo. After the i.v. injection of Adv-Ihh, the expression of vector-derived Ihh mRNA was detected in the liver. Four weeks after administration of Adv-Ihh to SCID mice, we observed an increase in the number of c-Kit+ cells and clonogenic cells per 10(5) mononuclear cells in the bone marrow compared with Adv-control-administered mice. Moreover, after administration of Adv-Ihh to BALB/c mice, the number of splenic B220+IgM(low)CD23(int)CD21(int) B lymphocytes and CD4+ T lymphocytes was strongly increased. Furthermore, the number of thymic double-negative (DN)2, DN3, CD8+ immature single-positive, and CD4+/CD8- cells was significantly elevated relative to the number in mice that received the control Adv vector. Our results suggest that enhanced signaling by Ihh can modulate the proliferation and differentiation of splenic B lymphocytes and thymic T lymphocytes during bone marrow hematopoiesis in vivo. Thus, modulation of the hedgehog signaling pathway may provide a therapeutic strategy to stimulate lymphomyelopoiesis in vivo.


Asunto(s)
Adenoviridae/genética , Proteínas Hedgehog/genética , Linfopoyesis/genética , Mielopoyesis/genética , Animales , Secuencia de Bases , Ensayo de Unidades Formadoras de Colonias , Cartilla de ADN/genética , Expresión Génica , Vectores Genéticos , Proteínas Hedgehog/fisiología , Humanos , Linfopoyesis/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Mielopoyesis/fisiología , Células 3T3 NIH , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Bazo/citología , Bazo/metabolismo , Transducción Genética
17.
Clin Cancer Res ; 14(1): 48-54, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18172251

RESUMEN

PURPOSE: Long-standing ulcerative colitis (UC) predisposes patients to the development of colorectal cancer, but surveillance of colitis-associated cancer by detecting the precancerous lesion dysplasia is often difficult because of its rare occurrence and normal-looking appearance. In sporadic colorectal cancer, aberrant crypt foci (ACF) have been reported by many investigators to be precursor lesions of the adenoma-carcinoma sequence. In the present study, we analyzed the genetic background of ACF to determine whether they could be precursors for dysplasia, and we examined the usefulness of endoscopic examination of ACF as a surrogate marker for surveillance of colitis-associated cancer. EXPERIMENTAL DESIGN: ACF were examined in 28 UC patients (19 patients with UC alone and 9 patients with UC and dysplasia; 2 of those patients with dysplasia also had cancer) using magnifying endoscopy. K-ras, APC, and p53 mutations were analyzed by two-step PCR RFLP, in vitro--synthesized protein assay, and single-strand conformation polymorphism, respectively. Methylation of p16 was analyzed by methylation-specific PCR. RESULTS: ACF that appeared distinct endoscopically and histologically were identified in 27 out of 28 UC patients. They were negative for K-ras, APC, and p53 mutations but were frequently positive for p16 methylation (8 of 11; 73%). In dysplasia, K-ras and APC mutations were negative but p53 mutation (3 of 5; 60%) and p16 methylation (3 of 5; 60%) were positive. There was a significant stepwise increase in the number of ACF from patients with UC alone to patients with dysplasia and to patients with cancer. Univariate and multivariate analyses showed significant correlations between ACF and dysplasia. CONCLUSIONS: We have disclosed an ACF-dysplasia-cancer sequence in colitis-associated carcinogenesis similar to the ACF-adenoma-carcinoma sequence in sporadic colon carcinogenesis. This study suggests the use of ACF instead of dysplasia for the surveillance of colitis cancer and warrants further evaluation of ACF as a surveillance marker in large-scale studies.


Asunto(s)
Colitis Ulcerosa/patología , Neoplasias Colorrectales/patología , Lesiones Precancerosas/patología , Adulto , Colitis Ulcerosa/genética , Neoplasias Colorrectales/genética , Progresión de la Enfermedad , Endoscopía Gastrointestinal , Femenino , Genes APC , Genes p16 , Genes p53 , Genes ras , Humanos , Masculino , Mutación , Polimorfismo de Longitud del Fragmento de Restricción , Polimorfismo Conformacional Retorcido-Simple , Lesiones Precancerosas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Exp Hematol ; 36(9): 1121-31, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18562079

RESUMEN

OBJECTIVE: Transplantation of hematopoietic stem cells (HSCs) is usually accomplished through intravenous injection, a complex process that requires recognition of bone marrow vasculature and migration to a supportive microenvironment. Hence, some populations of HSCs, including cord blood (CB) Lin(-)CD34(-) stem cells, do not engraft well in bone marrow (BM) of nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice. In this study, we examined the effect of human stromal interactions on the properties of CB Lin(-)CD34(-) cells. MATERIALS AND METHODS: CD34 and CXCR4 expression on fresh CB Lin(-)CD34(-) cells and CB Lin(-)CD34(-) cells cocultured with human stromal cells were analyzed. Homing activity and engraftment of these cells were assessed using NOD/SCID mice. In an attempt to identify the stromal CXCR4-inducing factor, CB Lin(-)CD34(-) cells were cocultured with a noncontact culture system in the presence of several inhibitors. RESULT: Coculture with human stromal cells induced expression of CD34 and CXCR4 on CB Lin(-)CD34(-) cells. CXCR4 expression on CB Lin(-)CD34(-) cells was induced even in the noncontact culture condition, suggesting that this CXCR4-inducing factor is soluble. Moreover, CXCR4 induction was inhibited by the soluble Wnt inhibitor DKK1. Furthermore, these cells acquired homing activity and engrafted in the BM of NOD/SCID mice after intravenous injection. CONCLUSION: These findings may be useful for understanding the role of stromal cells in homing and engraftment of HSCs.


Asunto(s)
Sangre Fetal/citología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/fisiología , Receptores CXCR4/biosíntesis , Células del Estroma/fisiología , Animales , Antígenos CD34/análisis , Antígenos de Diferenciación/análisis , Médula Ósea , Comunicación Celular , Quimiotaxis de Leucocito/fisiología , Técnicas de Cocultivo , Supervivencia de Injerto , Células Madre Hematopoyéticas/química , Células Madre Hematopoyéticas/clasificación , Humanos , Péptidos y Proteínas de Señalización Intercelular/fisiología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Receptores CXCR4/genética , Solubilidad , Trasplante Heterólogo , Regulación hacia Arriba/fisiología , Proteínas Wnt/antagonistas & inhibidores , Proteínas Wnt/biosíntesis , Proteínas Wnt/genética
19.
Int J Hematol ; 88(1): 30-35, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18581199

RESUMEN

Many patients with bone marrow failure syndromes need frequent transfusions of red blood cells, and most of them eventually suffer from organ dysfunction induced by excessively accumulated iron. The only way to treat transfusion-induced iron overload is iron chelating therapy. However, most patients have not been treated effectively because daily/continuous administration of deferoxamine is difficult for outpatients. Recently, a novel oral iron chelator, deferasirox, has been developed, and introduction of the drug may help many patients benefit from iron chelation therapy. In this review, we will discuss the current status of iron overload in transfusion-dependent patients, and the development of Japanese guidelines for the treatment of iron overload in Japan, which were established by the National Research Group on Idiopathic Bone Marrow Failure Syndromes in Japan.


Asunto(s)
Benzoatos/uso terapéutico , Enfermedades de la Médula Ósea/terapia , Recolección de Datos , Quelantes del Hierro/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/epidemiología , Guías de Práctica Clínica como Asunto , Triazoles/uso terapéutico , Pueblo Asiatico , Enfermedades de la Médula Ósea/complicaciones , Enfermedades de la Médula Ósea/epidemiología , Deferasirox , Transfusión de Eritrocitos/efectos adversos , Humanos , Sobrecarga de Hierro/etiología , Japón , Síndrome
20.
Int J Hematol ; 87(4): 339-350, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18369691

RESUMEN

We generated red blood cells (RBC) from cord blood (CB) CD34+ cells using a four-phase culture system. We first cultured CB CD34+ cells on telomerase gene-transduced human stromal cells in serum-free medium containing stem cell factor (SCF), Flt-3/Flk-2 ligand, and thrombopoietin to expand CD34+ cells (980-fold) and the total cells (10,400-fold) (first phase). Expanded cells from the first phase were liquid-cultured with SCF, interleukin-3 (IL-3), and erythropoietin (EPO) to expand (113-fold) and differentiate them into erythroblasts (second phase). To obtain macrophages for the next phase, we expanded CD34+ cells from a different donor using the same coculture system. Expanded cells from the first phase were liquid-cultured with granulocyte-macrophage colony stimulating factor, macrophage-colony stimulating factor (M-CSF), IL-3, and SCF to generate monocytes/macrophages (75-fold), which were incubated with type AB serum and M-CSF to fully differentiate them into macrophages. Erythroblasts were then co-cultured with macrophages in the presence of EPO to expand (threefold) and fully differentiate them (61% orthochromatic erythroblasts plus 39% RBC) (third phase). RBC were purified from erythroblasts and debris through a deleukocyting filter to generate 6.0 x 10(12) RBC from 1.0 unit of CB (3.0 transfusable units). Qualitatively, these RBC showed a hemoglobin content, oxygenation of hemoglobin, and in vivo clearance similar to those of adult peripheral RBC. Finally, an almost complete enucleation of orthochromatic erythroblasts (99.4%) was achieved by the cultivation method recently described by Miharada et al. in the absence of macrophages and cytokines (fourth phase). RBC were purified from remnant erythroblasts and debris by passage through a deleukocyting filter to generate 1.76 x 10(13) RBC from 1.0 unit of CB (8.8 transfusable units), the highest yield ever reported. Thus, this method may be useful for generating an alternative RBC supply for transfusions, investigating infectious agents that target erythroid cells, and as a general in vitro hematopoietic model system.


Asunto(s)
Antígenos CD34/metabolismo , Separación Celular/métodos , Eritrocitos/metabolismo , Sangre Fetal/metabolismo , Macrófagos/metabolismo , Diferenciación Celular , Técnicas de Cocultivo , Eritroblastos/citología , Eritroblastos/metabolismo , Eritrocitos/citología , Sangre Fetal/citología , Humanos , Macrófagos/citología , Telomerasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA