Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Leukemia ; 38(7): 1570-1580, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38454120

RESUMEN

Although Bruton's tyrosine kinase (BTK) inhibitors (BTKi) have significantly improved patient prognosis, mantle cell lymphoma (MCL) is still considered incurable due to primary and acquired resistance. We have recently shown that aberrant expression of the Src-family tyrosine kinase hematopoietic cell kinase (HCK) in MCL correlates with poor prognosis, and that genetic HCK perturbation impairs growth and integrin-mediated adhesion of MCL cells. Here, we show that KIN-8194, a dual inhibitor of BTK and HCK with in vivo activity against Myd88-L265P-driven diffuse large B-cell lymphoma and Waldenström Macroglobulinemia, has a potent growth inhibitory effect in MCL cell lines and primary MCL cells, irrespective of their sensitivity to BTKi (ibrutinib and acalabrutinib). In BTKi-resistant cells this is mediated by inhibition of HCK, which results in repression of AKT-S6 signaling. In addition, KIN-8194 inhibits integrin-mediated adhesion of BTKi-sensitive and insensitive MCL cells to fibronectin and stromal cells in an HCK-dependent manner. Finally, we show that MCL cells with acquired BTKi resistance retain their sensitivity to KIN-8194. Taken together, our data demonstrate that KIN-8194 inhibits growth and integrin-mediated adhesion of BTKi-sensitive MCL cells, as well as MCL cells with primary or acquired BTKi resistance. This renders KIN-8194 a promising novel treatment for MCL patients.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa , Adhesión Celular , Proliferación Celular , Resistencia a Antineoplásicos , Integrinas , Linfoma de Células del Manto , Inhibidores de Proteínas Quinasas , Proteínas Proto-Oncogénicas c-hck , Linfoma de Células del Manto/tratamiento farmacológico , Linfoma de Células del Manto/patología , Linfoma de Células del Manto/metabolismo , Linfoma de Células del Manto/genética , Humanos , Adhesión Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-hck/metabolismo , Proteínas Proto-Oncogénicas c-hck/genética , Proteínas Proto-Oncogénicas c-hck/antagonistas & inhibidores , Integrinas/metabolismo , Proliferación Celular/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Línea Celular Tumoral , Pirimidinas/farmacología , Adenina/análogos & derivados , Adenina/farmacología , Transducción de Señal/efectos de los fármacos
3.
J Hematol Oncol ; 14(1): 11, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33436043

RESUMEN

BACKGROUND: The survival and proliferation of multiple myeloma (MM) cells in the bone marrow (BM) critically depend on interaction with stromal cells expressing the chemokine CXCL12. CXCL12 regulates the homing to the BM niche by mediating the transendothelial migration and adhesion/retention of the MM cells. The gamma isoform of CXCL12 (CXCL12γ) has been reported to be highly expressed in mouse BM and to show enhanced biological activity compared to the 'common' CXCL12α isoform, mediated by its unique extended C-terminal domain, which binds heparan sulfate proteoglycans (HSPGs) with an extraordinary high affinity. Here, we investigated the expression of CXCL12γ in human BM and studied its functional role in the interaction of MM cells with BM stromal cells (BMSCs). METHODS: We assessed CXCL12γ mRNA and protein expression by human BMSCs using qPCR, flow cytometry, and immunohistochemistry. CRISPR-Cas9 was employed to delete CXCL12γ and the heparan sulfate (HS) co-polymerase EXT1 in BMSCs. To study the functional roles of BMSC-derived CXCL12γ and HSPGs in the interaction of MM cells with BMSCs cells, MM cell lines and primary MM cells were co-cultured with BMSCs. RESULTS: We observed that CXCL12γ is expressed in situ by reticular stromal cells in both normal and MM BM, as well as by primary BMSC isolates and BMSC lines. Importantly, upon secretion, CXCL12γ, unlike the CXCL12α isoform, was retained on the surface of BMSCs. This membrane retention of CXCL12γ is HSPG mediated, since it was completely annulated by CRISPR-Cas9-mediated deletion of the HS co-polymerase EXT1. CXCL12γ expressed by BMSCs and membrane-retained by HSPGs supported robust adhesion of MM cells to the BMSCs. Specific genetic deletion of either CXCL12γ or EXT1 significantly attenuated the ability of BMSCs to support MM cell adhesion and, in addition, impaired their capacity to protect MM cells from bortezomib-induced cell death. CONCLUSIONS: We show that CXCL12γ is expressed by human BMSCs and upon secretion is retained on their cell surface by HSPGs. The membrane-bound CXCL12γ controls adhesion of MM cells to the stromal niche and mediates drug resistance. These findings designate CXCL12γ and associated HSPGs as partners in mediating MM-niche interaction and as potential therapeutic targets in MM.


Asunto(s)
Adhesión Celular , Quimiocina CXCL12/metabolismo , Heparitina Sulfato/metabolismo , Mieloma Múltiple/metabolismo , Adhesión Celular/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Técnicas de Cocultivo , Resistencia a Antineoplásicos , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Mieloma Múltiple/tratamiento farmacológico , Inhibidores de Proteasoma/farmacología
4.
Leukemia ; 35(3): 881-886, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32591642

RESUMEN

Mantle cell lymphoma (MCL) is an aggressive non-Hodgkin lymphoma subtype arising from naïve B cells. Although novel therapeutics have improved patient prognosis, drug resistance remains a key problem. Here, we show that the SRC-family tyrosine kinase hematopoietic cell kinase (HCK), which is primarily expressed in the hematopoietic lineage but not in mature B cells, is aberrantly expressed in MCL, and that high expression of HCK is associated with inferior prognosis of MCL patients. HCK expression is controlled by the toll-like receptor (TLR) adaptor protein MYD88 and can be enhanced by TLR agonists in MCL cell lines and primary MCL. In line with this, primary MCL with high HCK expression are enriched for a TLR-signaling pathway gene set. Silencing of HCK expression results in cell cycle arrest and apoptosis. Furthermore, HCK controls integrin-mediated adhesion of MCL cells to extracellular matrix and stromal cells. Taken together, our data indicate that TLR/MYD88-controlled aberrant expression of HCK plays a critical role in MCL proliferation and survival as well as in retention of the malignant cells in the growth- and survival-supporting lymphoid organ microenvironment, thereby contributing to lymphomagenesis. These novel insights provide a strong rationale for therapeutic targeting of HCK in MCL.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Linfoma de Células del Manto/patología , Proteínas Proto-Oncogénicas c-hck/metabolismo , Microambiente Tumoral , Biomarcadores de Tumor/genética , Humanos , Linfoma de Células del Manto/genética , Linfoma de Células del Manto/metabolismo , Pronóstico , Proteínas Proto-Oncogénicas c-hck/genética , Transducción de Señal
5.
Oncotarget ; 8(42): 71981-71995, 2017 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-29069762

RESUMEN

Chronic lymphocytic leukemia (CLL) is characterized by the accumulation of mature CD5+ B cells in blood. Spontaneous apoptosis of CLL cells in vitro has hampered in-depth investigation of CLL pathogenesis. Here we describe the generation of three monoclonal mouse cell lines, EMC2, EMC4 and EMC6, from the IgH.TEµ CLL mouse model based on sporadic expression of SV40 large T antigen. The cell lines exhibit a stable CD5+CD43+IgM+CD19+ CLL phenotype in culture and can be adoptively transferred into Rag1-/- mice. RNA-seq analysis revealed only minor differences between the cell lines and their primary tumors and suggested that NF-κB and mTOR signaling pathways were involved in cell line outgrowth. In vitro survival and proliferation was dependent on constitutive phosphorylation of Bruton's tyrosine kinase (Btk) at Y551/Y223, and Akt(S473). Treatment of the cell lines with small molecule inhibitors specific for Btk (ibrutinib) or PI3K (idelalisib), which is upstream of Akt, resulted in reduced viability, proliferation and fibronectin-dependent cell adhesion. Treatment of cell line-engrafted Rag1-/- mice with ibrutinib was associated with transient lymphocytosis, reduced splenomegaly and increased overall survival. Thus, by generating stable cell lines we established a novel platform for in vitro and in vivo investigation of CLL signal transduction and treatment modalities.

6.
Haematologica ; 101(3): e111-5, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26635033
8.
Blood ; 122(14): 2412-24, 2013 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-23940282

RESUMEN

Ibrutinib (PCI-32765) is a highly potent oral Bruton tyrosine kinase (BTK) inhibitor in clinical development for treating B-cell lymphoproliferative diseases. Patients with chronic lymphocytic leukemia (CLL) often show marked, transient increases of circulating CLL cells following ibrutinib treatments, as seen with other inhibitors of the B-cell receptor (BCR) pathway. In a phase 1 study of ibrutinib, we noted similar effects in patients with mantle cell lymphoma (MCL). Here, we characterize the patterns and phenotypes of cells mobilized among patients with MCL and further investigate the mechanism of this effect. Peripheral blood CD19(+)CD5(+) cells from MCL patients were found to have significant reduction in the expression of CXCR4, CD38, and Ki67 after 7 days of treatment. In addition, plasma chemokines such as CCL22, CCL4, and CXCL13 were reduced 40% to 60% after treatment. Mechanistically, ibrutinib inhibited BCR- and chemokine-mediated adhesion and chemotaxis of MCL cell lines and dose-dependently inhibited BCR, stromal cell, and CXCL12/CXCL13 stimulations of pBTK, pPLCγ2, pERK, or pAKT. Importantly, ibrutinib inhibited migration of MCL cells beneath stromal cells in coculture. We propose that BTK is essential for the homing of MCL cells into lymphoid tissues, and its inhibition results in an egress of malignant cells into peripheral blood. This trial was registered at www.clinicaltrials.gov as #NCT00114738.


Asunto(s)
Antineoplásicos/uso terapéutico , Linfocitos B/efectos de los fármacos , Quimiotaxis de Leucocito/efectos de los fármacos , Linfoma de Células del Manto/sangre , Pirazoles/uso terapéutico , Pirimidinas/uso terapéutico , Adenina/análogos & derivados , Antígenos CD19/biosíntesis , Linfocitos B/metabolismo , Western Blotting , Antígenos CD5/biosíntesis , Adhesión Celular/efectos de los fármacos , Citometría de Flujo , Humanos , Linfoma de Células del Manto/tratamiento farmacológico , Piperidinas , Inhibidores de Proteínas Quinasas/uso terapéutico
9.
Blood ; 119(11): 2590-4, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22279054

RESUMEN

Small-molecule drugs that target the B-cell antigen receptor (BCR) signalosome show clinical efficacy in the treatment of B-cell non-Hodgkin lymphoma. These agents, including the Bruton tyrosine kinase (BTK) inhibitor PCI-32765, display an unexpected response in patients with chronic lymphocytic leukemia (CLL): a rapid and sustained reduction of lymphadenopathy accompanied by transient lymphocytosis, which is reversible upon temporary drug deprivation. We hypothesized that this clinical response reflects impaired integrin-mediated adhesion and/or migration. Here, we show that PCI-32765 strongly inhibits BCR-controlled signaling and integrin α(4)ß(1)-mediated adhesion to fibronectin and VCAM-1 of lymphoma cell lines and primary CLL cells. Furthermore, PCI-32765 also inhibits CXCL12-, CXCL13-, and CCL19-induced signaling, adhesion, and migration of primary CLL cells. Our data indicate that inhibition of BTK by PCI-32765 overcomes BCR- and chemokine-controlled integrin-mediated retention and homing of malignant B cells in their growth- and survival-supporting lymph node and bone marrow microenvironment, which results in clinically evident CLL regression.


Asunto(s)
Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Quimiocinas/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirazoles/farmacología , Pirimidinas/farmacología , Receptores de Antígenos de Linfocitos B/metabolismo , Adenina/análogos & derivados , Agammaglobulinemia Tirosina Quinasa , Apoptosis/efectos de los fármacos , Western Blotting , Proliferación Celular/efectos de los fármacos , Fibronectinas/metabolismo , Citometría de Flujo , Humanos , Integrina alfa4beta1/metabolismo , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/metabolismo , Piperidinas , Proteínas Tirosina Quinasas/metabolismo , ARN Mensajero/genética , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas , Molécula 1 de Adhesión Celular Vascular/metabolismo
10.
Blood ; 117(23): 6162-71, 2011 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-21471524

RESUMEN

The development and antigen-dependent differentiation of B lymphocytes are orchestrated by an array of growth factors, cytokines, and chemokines that require tight spatiotemporal regulation. Heparan sulfate proteoglycans specifically bind and regulate the bioavailability of soluble protein ligands, but their role in the immune system has remained largely unexplored. Modification of heparan sulfate by glucuronyl C5-epimerase (Glce) controls heparan sulfate-chain flexibility and thereby affects ligand binding. Here we show that Glce deficiency impairs B-cell maturation, resulting in decreased plasma cell numbers and immunoglobulin levels. We demonstrate that C5-epimerase modification of heparan sulfate is critical for binding of a proliferation inducing ligand (APRIL) and that Glce-deficient plasma cells fail to respond to APRIL-mediated survival signals. Our results identify heparan sulfate proteoglycans as novel players in B-cell maturation and differentiation and suggest that heparan sulfate conformation is crucial for recruitment of factors that control plasma cell survival.


Asunto(s)
Carbohidrato Epimerasas/inmunología , Diferenciación Celular/inmunología , Proteoglicanos de Heparán Sulfato/inmunología , Células Plasmáticas/inmunología , Transducción de Señal/inmunología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Animales , Carbohidrato Epimerasas/genética , Diferenciación Celular/genética , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Proteoglicanos de Heparán Sulfato/genética , Proteoglicanos de Heparán Sulfato/metabolismo , Ratones , Ratones Noqueados , Células Plasmáticas/enzimología , Transducción de Señal/genética , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo
11.
J Immunol ; 184(7): 3656-64, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20208005

RESUMEN

The development of lymphoid organs depends on cross talk between hematopoietic cells and mesenchymal stromal cells and on vascularization of the lymphoid primordia. These processes are orchestrated by cytokines, chemokines, and angiogenic factors that require tight spatiotemporal regulation. Heparan sulfate (HS) proteoglycans are molecules designed to specifically bind and regulate the bioactivity of soluble protein ligands. Their binding capacity and specificity are controlled by modification of the HS side chain by HS-modifying enzymes. Although HS proteoglycans have been implicated in the morphogenesis of several organ systems, their role in controlling lymphoid organ development has thus far remained unexplored. In this study, we report that modification of HS by the HS-modifying enzyme glucuronyl C5-epimerase (Glce), which controls HS chain flexibility, is required for proper lymphoid organ development. Glce(-/-) mice show a strongly reduced size of the fetal spleen as well as a spectrum of defects in thymus and lymph node development, ranging from dislocation to complete absence of the organ anlage. Once established, however, the Glce(-/-) primordia recruited lymphocytes and developed normal architectural features. Furthermore, Glce(-/-) lymph node anlagen transplanted into wild-type recipient mice allowed undisturbed lymphocyte maturation. Our results indicate that modification of HS by Glce is required for controlling the activity of molecules that are instructive for early lymphoid tissue morphogenesis but may be dispensable at later developmental stages and for lymphocyte maturation and differentiation.


Asunto(s)
Carbohidrato Epimerasas/inmunología , Proteoglicanos de Heparán Sulfato/metabolismo , Tejido Linfoide/embriología , Tejido Linfoide/enzimología , Organogénesis/inmunología , Animales , Carbohidrato Epimerasas/deficiencia , Separación Celular , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Proteoglicanos de Heparán Sulfato/inmunología , Ratones , Ratones Noqueados
12.
Blood ; 115(3): 601-4, 2010 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-19965677

RESUMEN

Expression of the heparan sulfate proteoglycan syndecan-1 is a hallmark of both normal and multiple myeloma (MM) plasma cells. Syndecan-1 could affect plasma cell fate by strengthening integrin-mediated adhesion via its core protein and/or by accommodating and presenting soluble factors via its HS side chains. Here, we show that inducible RNAi-mediated knockdown of syndecan-1 in human MM cells leads to reduced growth rates and a strong increase of apoptosis. Importantly, knockdown of EXT1, a copolymerase critical for HS chain biosynthesis, had similar effects. Using an innovative myeloma xenotransplantation model in Rag-2(-/-)gamma(c)(-/-) mice, we demonstrate that induction of EXT1 knockdown in vivo dramatically suppresses the growth of bone marrow localized myeloma. Our findings provide direct evidence that the HS chains of syndecan-1 are crucial for the growth and survival of MM cells within the bone marrow environment, and indicate the HS biosynthesis machinery as a potential treatment target in MM.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Heparitina Sulfato/fisiología , Mieloma Múltiple/patología , N-Acetilglucosaminiltransferasas/genética , ARN Interferente Pequeño/farmacología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Doxiciclina/administración & dosificación , Sistemas de Liberación de Medicamentos , Marcación de Gen , Heparitina Sulfato/metabolismo , Humanos , Cadenas gamma de Inmunoglobulina/genética , Ratones , Ratones Noqueados , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , N-Acetilglucosaminiltransferasas/antagonistas & inhibidores , N-Acetilglucosaminiltransferasas/fisiología , Sindecano-1/genética , Sindecano-1/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Blood ; 111(7): 3364-72, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18227351

RESUMEN

Chemokine-controlled migration plays a critical role in B-cell development, differentiation, and function, as well as in the pathogenesis of B-cell malignancies, including the plasma cell neoplasm multiple myeloma (MM). Here, we demonstrate that stimulation of B cells and MM cells with the chemokine stromal cell-derived factor-1 (SDF-1) induces strong migration and activation of the Ras-like GTPase Ral. Inhibition of Ral, by expression of the dominant negative RalN28 mutant or of RalBPDeltaGAP, a Ral effector mutant that sequesters active Ral, results in impaired SDF-1-induced migration of B cells and MM cells. Of the 2 Ral isoforms, RalA and RalB, RalB was found to mediate SDF-1-induced migration. We have recently shown that Btk, PLCgamma2, and Lyn/Syk mediate SDF-1-controlled B-cell migration; however, SDF-1-induced Ral activation is not affected in B cells deficient in these proteins. In addition, treatment with pharmacological inhibitors against PI3K and PLC or expression of dominant-negative Ras did not impair SDF-1-induced Ral activation. Taken together, these results reveal a novel function for Ral, that is, regulation of SDF-1-induced migration of B cells and MM cells, thereby providing new insights into the control of B-cell homeostasis, trafficking, and function, as well as into the pathogenesis of MM.


Asunto(s)
Linfocitos B/metabolismo , Movimiento Celular , Quimiocina CXCL12/metabolismo , Leucemia de Células Plasmáticas/metabolismo , Proteínas de Unión al GTP ral/metabolismo , Agammaglobulinemia Tirosina Quinasa , Animales , Linfocitos B/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Quimiocina CXCL12/farmacología , Pollos , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Genes Dominantes/genética , Homeostasis/efectos de los fármacos , Homeostasis/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Leucemia de Células Plasmáticas/genética , Leucemia de Células Plasmáticas/patología , Mutación , Proteína Oncogénica p21(ras)/genética , Proteína Oncogénica p21(ras)/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfolipasa C gamma/genética , Fosfolipasa C gamma/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Quinasa Syk , Proteínas de Unión al GTP ral/genética , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
14.
Mol Pharmacol ; 72(2): 387-94, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17485564

RESUMEN

Glucuronidation is a major hepatic detoxification pathway for endogenous and exogenous compounds, resulting in the intracellular formation of polar metabolites that require specialized transporters for elimination. Multidrug resistance proteins (MRPs) are expressed in the liver and can transport glucuronosyl-conjugates. Using morphine as a model aglycone, we demonstrate that morphine-3-glucuronide (M3G), the predominant metabolite, is transported in vitro by human MRP2 (ABCC2), a protein present in the apical membrane of hepatocytes. Loss of biliary M3G secretion in Mrp2(-/-) mice results in its increased sinusoidal transport that can be attributed to Mrp3. Combined loss of Mrp2 and Mrp3 leads to a substantial accumulation of M3G in the liver, from which it is transported across the sinusoidal membrane at a low rate, resulting in the prolonged presence of M3G in plasma. Our results show that murine Mrp2 and Mrp3 provide alternative routes for the excretion of a glucuronidated substrate from the liver in vivo.


Asunto(s)
Hígado/metabolismo , Proteínas de Transporte de Membrana/fisiología , Derivados de la Morfina/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/fisiología , Animales , Línea Celular , Glucuronosiltransferasa/fisiología , Humanos , Ratones , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Spodoptera
15.
Mol Pharmacol ; 71(1): 240-9, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17032904

RESUMEN

Some cellular uptake systems for (anti)folates function optimally at acidic pH. We have tested whether this also applies to efflux from cells by breast cancer resistance protein (BCRP; ABCG2), which has been reported to transport folic acid, methotrexate, and methotrexate di- and triglutamate at physiological pH. Using Spodoptera frugiperda-BCRP membrane vesicles, we showed that the ATP-dependent vesicular transport of 1 muM methotrexate by BCRP is 5-fold higher at pH 5.5 than at physiological pH. The transport of methotrexate was saturable at pH 5.5, with apparent Km and Vmax values of 1.3 +/- 0.2 mM and 44 +/- 2.5 nmol/mg of protein/min, respectively, but was linear with drug concentration at pH 7.3 up to 6 mM methotrexate. In contrast to recent reports, we did not detect transport of methotrexate diglutamate at physiological pH, but we did find transport at pH 5.5. We also found that 7-hydroxy-methotrexate, the major metabolite of methotrexate, is transported by BCRP both at physiological pH and (more efficiently) at low pH. The pH effect was also observed in intact BCRP-overexpressing cells: we found a 3-fold higher level of resistance to both methotrexate and the prototypical BCRP substrate mitoxantrone at pH 6.5 as at physiological pH. Furthermore, with MDCKII-BCRP monolayers, we found that resveratrol, which is a neutral compound at pH < or = 7.4, is efficiently transported by BCRP at pH 6.0, whereas we did not detect active transport at pH 7.4. We conclude that BCRP transports substrate drugs more efficiently at low pH, independent of the dissociation status of the substrate.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Antineoplásicos/farmacocinética , Neoplasias de la Mama/metabolismo , Ácido Fólico/farmacocinética , Concentración de Iones de Hidrógeno , Metotrexato/análogos & derivados , Metotrexato/farmacocinética , Mitoxantrona/farmacocinética , Proteínas de Neoplasias/metabolismo , Estilbenos/farmacocinética , Topotecan/farmacocinética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Animales , Transporte Biológico , Línea Celular , Línea Celular Tumoral , Perros , Femenino , Humanos , Cinética , Hígado/metabolismo , Conejos , Resveratrol
16.
J Hepatol ; 44(4): 768-75, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16225954

RESUMEN

BACKGROUND/AIM: Multidrug Resistance Protein 3 (MRP3) transports bile salts and glucuronide conjugates in vitro and is postulated to protect the liver in cholestasis. Whether the absence of Mrp3 affects these processes in vivo is tested. METHODS: Mrp3-deficient mice were generated and the contribution of Mrp3 to bile salt and glucuronide conjugate transport was tested in (1): an Ussing-chamber set-up with ileal explants (2), the liver during bile-duct ligation (3), liver perfusion experiments, and (4) in vitro vesicular uptake experiments. RESULTS: The Mrp3((-/-)) mice show no overt phenotype. No differences between WT and Mrp3-deficient mice were found in the trans-ileal transport of taurocholate. After bile-duct ligation, there were no differences in histological liver damage and serum bile salt levels between Mrp3((-/-)) and WT mice, but Mrp3-deficient mice had lower serum bilirubin glucuronide concentrations. Glucuronide conjugates of hyocholate and hyodeoxycholate are substrates of MRP3 in vitro and in livers that lack Mrp3, there is reduced sinusoidal secretion of hyodeoxycholate-glucuronide after perfusion with hyodeoxycholate. CONCLUSIONS: Mrp3 does not have a major role in bile salt physiology, but is involved in the transport of glucuronidated compounds, which could include glucuronidated bile salts in humans.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Glucurónidos/metabolismo , Hígado/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/deficiencia , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/fisiología , Animales , Conductos Biliares/fisiopatología , Bilirrubina/análogos & derivados , Bilirrubina/sangre , Transporte Biológico/genética , Transporte Biológico/fisiología , Ácidos Cólicos/metabolismo , Ácido Desoxicólico/metabolismo , Íleon/metabolismo , Immunoblotting , Inmunohistoquímica , Ligadura , Hígado/química , Masculino , Ratones , Ratones Endogámicos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/análisis , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Ácido Taurocólico/metabolismo
17.
Clin Cancer Res ; 11(12): 4487-94, 2005 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15958634

RESUMEN

Multidrug resistance (MDR) remains a major obstacle to successful chemotherapeutic treatment of cancer and can be caused by overexpression of P-glycoprotein, the MDR1 gene product. To further validate a knockdown approach for circumventing MDR, we developed a P-glycoprotein inhibition strategy using short hairpin RNA interference (shRNAi) and now show efficacy and target specificity in vivo. Two of eight tested shRNAi constructs targeted against human MDR1 mRNA inhibited expression of P-glycoprotein by >90%, whereas control shRNAi had no effect. Ablation of P-glycoprotein in cells stably transduced with retroviral-mediated shRNAi was documented by Western blot and functionally confirmed by increased sensitivity of MDR1-transfected cells toward the cytotoxic drugs vincristine, paclitaxel, and doxorubicin as well as by transport of (99m)Tc-Sestamibi. shRNAi-mediated down-regulation of P-glycoprotein transport activity both in cultured cells and in tumor implants in living animals could be followed by direct noninvasive bioluminescence imaging using the Renilla luciferase fluorophore, coelenterazine, a known P-glycoprotein transport substrate. Furthermore, after somatic gene transfer by hydrodynamic infusion of a MDR1-Firefly luciferase (MDR1-FLuc) fusion construct into mouse liver, the effect of shRNAi delivered in vivo on P-glycoprotein-FLuc protein levels was documented with bioluminescence imaging using d-luciferin. ShRNAi against MDR1 reduced bioluminescence output of the P-glycoprotein-FLuc reporter 4-fold in vivo compared with mice treated with control or scrambled shRNAi. Targeted down-regulation of a somatically transferred P-glycoprotein-eGFP fusion reporter also was observed using fluorescence microscopy. Our results show that shRNAi effectively inhibited MDR1 expression and function in cultured cells, tumor implants and mammalian liver, documenting the feasibility of a knockdown approach to reversing MDR in vivo.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Interferencia de ARN , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Transporte Biológico , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Immunoblotting , Luciferasas/genética , Luciferasas/metabolismo , Mediciones Luminiscentes , Masculino , Ratones , Ratones Desnudos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Vincristina/farmacología
18.
Proc Natl Acad Sci U S A ; 102(20): 7274-9, 2005 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-15886284

RESUMEN

Glucuronidation is a major detoxification pathway for endogenous and exogenous compounds in mammals that results in the intracellular formation of polar metabolites, requiring specialized transporters to cross biological membranes. By using morphine as a model aglycone, we demonstrate that multidrug resistance protein 3 (MRP3/ABCC3), a protein present in the basolateral membrane of polarized cells, transports morphine-3-glucuronide (M3G) and morphine-6-glucuronide in vitro. Mrp3(-/-) mice are unable to excrete M3G from the liver into the bloodstream, the major hepatic elimination route for this drug. This results in increased levels of M3G in liver and bile, a 50-fold reduction in the plasma levels of M3G, and in a major shift in the main disposition route for morphine and M3G, predominantly via the urine in WT mice but via the feces in Mrp3(-/-) mice. The pharamacokinetics of injected morphine-glucuronides are altered as well in the absence of Mrp3, and this results in a decreased antinociceptive potency of injected morphine-6-glucuronide.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Derivados de la Morfina/metabolismo , Morfina/farmacocinética , Subfamilia B de Transportador de Casetes de Unión a ATP/deficiencia , Animales , Bilis/metabolismo , Línea Celular , Glucuronosiltransferasa , Humanos , Hígado/metabolismo , Ratones , Ratones Noqueados , Derivados de la Morfina/sangre , Derivados de la Morfina/farmacocinética , Derivados de la Morfina/farmacología , Dimensión del Dolor/efectos de los fármacos , Transporte de Proteínas , Spodoptera , Distribución Tisular , Vesículas Transportadoras/metabolismo
19.
Proc Natl Acad Sci U S A ; 100(16): 9244-9, 2003 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-12835412

RESUMEN

Prostaglandins are involved in a wide variety of physiological and pathophysiological processes, but the mechanism of prostaglandin release from cells is not completely understood. Although poorly membrane permeable, prostaglandins are believed to exit cells by passive diffusion. We have investigated the interaction between prostaglandins and members of the ATP-binding cassette (ABC) transporter ABCC [multidrug resistance protein (MRP)] family of membrane export pumps. In inside-out membrane vesicles derived from insect cells or HEK293 cells, MRP4 catalyzed the time- and ATP-dependent uptake of prostaglandin E1 (PGE1) and PGE2. In contrast, MRP1, MRP2, MRP3, and MRP5 did not transport PGE1 or PGE2. The MRP4-mediated transport of PGE1 and PGE2 displayed saturation kinetics, with Km values of 2.1 and 3.4 microM, respectively. Further studies showed that PGF1alpha, PGF2alpha, PGA1, and thromboxane B2 were high-affinity inhibitors (and therefore presumably substrates) of MRP4. Furthermore, several nonsteroidal antiinflammatory drugs were potent inhibitors of MRP4 at concentrations that did not inhibit MRP1. In cells expressing the prostaglandin transporter PGT, the steady-state accumulation of PGE1 and PGE2 was reduced proportional to MRP4 expression. Inhibition of MRP4 by an MRP4-specific RNA interference construct or by indomethacin reversed this accumulation deficit. Together, these data suggest that MRP4 can release prostaglandins from cells, and that, in addition to inhibiting prostaglandin synthesis, some nonsteroidal antiinflammatory drugs might also act by inhibiting this release.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Proteínas Asociadas a Resistencia a Múltiples Medicamentos , Prostaglandinas/metabolismo , Proteínas Ribosómicas/química , Proteínas Ribosómicas/fisiología , Adenosina Trifosfato/metabolismo , Alprostadil/metabolismo , Animales , Transporte Biológico , Línea Celular , Membrana Celular/metabolismo , Difusión , Dinoprost/metabolismo , Dinoprostona/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Insectos , Prostaglandinas A/metabolismo , Prostaglandinas F/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Tromboxano B2/metabolismo , Factores de Tiempo
20.
J Biol Chem ; 278(26): 23538-44, 2003 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-12702717

RESUMEN

Multidrug resistance protein 2 (MRP2) belongs to the ATP binding cassette family of transporters. Its substrates include organic anions and anticancer drugs. We have used transport assays with vesicles derived from Sf9 insect cells overproducing MRP2 to study the interactions of drugs, organic anions, and bile acids with three MRP2 substrates: estradiol-17-beta-d-glucuronide (E217betaG), methotrexate, and glutathione-S-dinitrophenol. Complex inhibition and stimulation patterns were obtained, different from those observed with the related transporters MRP1 and MRP3. In contrast to a previous report, we found that the rate of E217betaG transport by MRP2 increases sigmoidally with substrate concentration indicative of homotropic cooperativity. Half-maximal transport was obtained at 120 microm E217betaG, in contrast to values < 20 microm for MRP1 and 3. MRP2 stimulators, such as indomethacin and sulfanitran, strongly increased the affinity of MRP2 for E217betaG (half-maximal transport rates at 65 and 16 microm E217betaG, respectively) and shifted the sigmoidal dependence of transport rate on substrate concentration to a more hyperbolic one, without substantially affecting the maximal transport rate. Sulfanitran also stimulated MRP2 activity in cells, i.e. the transport of saquinavir through monolayers of Madin-Darby canine kidney II cells. Some compounds that stimulate E217betaG transport, such as penicillin G or pantoprazole, are not detectably transported by MRP2, suggesting that they allosterically stimulate transport without being cotransported with E217betaG. We propose that MRP2 contains two similar but nonidentical ligand binding sites: one site from which substrate is transported and a second site that regulates the affinity of the transport site for the substrate.


Asunto(s)
Regulación Alostérica , Estradiol/análogos & derivados , Proteínas Mitocondriales , Proteínas Ribosómicas/química , Proteínas Ribosómicas/metabolismo , Proteínas de Saccharomyces cerevisiae , Animales , Sitios de Unión , Transporte Biológico , Línea Celular , Estradiol/metabolismo , Humanos , Cinética , Ligandos , Metotrexato/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Transducción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA