Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Pediatr Rheumatol Online J ; 21(1): 65, 2023 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-37391782

RESUMEN

OBJECTIVE: Chronic nonbacterial osteomyelitis (CNO) is an autoinflammatory bone disorder that predominantly affects children and young people. The pathophysiology and molecular mechanisms of CNO remain poorly understood, and diagnostic criteria and biomarkers are lacking. As a result, treatment is empiric and follows personal experience, case series and expert consensus plans. METHODS: A survey was designed to gain insight on clinician and patient experiences of diagnosing and treating CNO and to collate opinions on research priorities. A version containing 24 questions was circulated among international expert clinicians and clinical academics (27 contacted, 21 responses). An equivalent questionnaire containing 20 questions was shared to explore the experience and priorities of CNO patients and family members (93 responses). RESULTS: Responses were used to select topics for four moderated roundtable discussions at the "International Conference on CNO and autoinflammatory bone disease" (Liverpool, United Kingdom, May 25-26th, 2022). The group identified deciphering the pathophysiology of CNO to be the highest priority, followed by clinical trials, necessary outcome measures and classification criteria. Surprisingly, mental wellbeing scored behind these items. CONCLUSIONS: Agreement exists among clinicians, academics, patients and families that deciphering the pathophysiology of CNO is of highest priority to inform clinical trials that will allow for the approval of medications for the treatment of CNO by regulatory agencies.


Asunto(s)
Osteomielitis , Adolescente , Niño , Humanos , Enfermedades Óseas , Consenso , Osteomielitis/diagnóstico , Osteomielitis/terapia
2.
Clin Immunol ; 251: 109344, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37098355

RESUMEN

Chronic nonbacterial osteomyelitis (CNO) is an autoinflammatory bone disease that primarily affects children and adolescents. CNO is associated with pain, bone swelling, deformity, and fractures. Its pathophysiology is characterized by increased inflammasome assembly and imbalanced expression of cytokines. Treatment is currently based on personal experience, case series and resulting expert recommendations. Randomized controlled trials (RCTs) have not been initiated because of the rarity of CNO, expired patent protection of some medications, and the absence of agreed outcome measures. An international group of fourteen CNO experts and two patient/parent representatives was assembled to generate consensus to inform and conduct future RCTs. The exercise delivered consensus inclusion and exclusion criteria, patent protected (excludes TNF inhibitors) treatments of immediate interest (biological DMARDs targeting IL-1 and IL-17), primary (improvement of pain; physician global assessment) and secondary endpoints (improved MRI; improved PedCNO score which includes physician and patient global scores) for future RCTs in CNO.


Asunto(s)
Antirreumáticos , Osteomielitis , Niño , Adolescente , Humanos , Consenso , Citocinas , Antirreumáticos/uso terapéutico , Osteomielitis/tratamiento farmacológico , Dolor/complicaciones , Dolor/tratamiento farmacológico , Enfermedad Crónica
3.
PLoS One ; 14(6): e0215739, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31166977

RESUMEN

INTRODUCTION: Chronic nonbacterial osteomyelitis (CNO) is a rare autoinflammatory bone disorder primarily affecting children and adolescents. It can lead to chronic pain, bony deformities and fractures. The pathophysiology of CNO is incompletely understood. Scientific evidence suggests dysregulated expression of pro- and anti-inflammatory cytokines to be centrally involved. Currently, treatment is largely based on retrospective observational studies and expert opinion. Treatment usually includes nonsteroidal anti-inflammatory drugs and/or glucocorticoids, followed by a range of drugs in unresponsive cases. While randomised clinical trials are lacking, retrospective and prospective non-controlled studies suggest effectiveness of TNF inhibitors and bisphosphonates. The objective of the Bayesian consensus meeting was to quantify prior expert opinion. METHODS: Twelve international CNO experts were randomly chosen to be invited to a Bayesian prior elicitation meeting. RESULTS: Results showed that a typical new patient treated with pamidronate would have an 84% chance of improvement in their pain score relative to baseline at 26 weeks and an 83% chance on adalimumab. Experts thought there was a 50% chance that a new typical patient would record a pain score of 28mm (pamidronate) to 30mm (adalimumab) or better at 26 weeks. There was a modest trend in prior opinion to indicate an advantage of pamidronate vs adalimumab, with a 68% prior chance that pamidronate is superior to adalimumab by some margin. However, it is clear that there is considerable uncertainty about the precise relative merits of the two treatments. CONCLUSIONS: The rarity of CNO leads to challenges in conducting randomised controlled trials with sufficient power to provide a definitive outcome. We address this using a Bayesian design, and here describe the process and outcome of the elicitation exercise to establish expert prior opinion. This opinion will be tested in the planned prospective CNO study. The process for establishing expert consensus opinion in CNO will be helpful for developing studies in other rare paediatric diseases.


Asunto(s)
Adalimumab/uso terapéutico , Osteomielitis/tratamiento farmacológico , Pamidronato/uso terapéutico , Teorema de Bayes , Consenso , Femenino , Humanos , Masculino , Osteomielitis/complicaciones , Manejo del Dolor , Ensayos Clínicos Controlados Aleatorios como Asunto , Proyectos de Investigación
5.
Mol Psychiatry ; 18(10): 1077-89, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23711981

RESUMEN

Autism spectrum disorders (ASDs) have been suggested to arise from abnormalities in the canonical and non-canonical Wnt signaling pathways. However, a direct connection between a human variant in a Wnt pathway gene and ASD-relevant brain pathology has not been established. Prickle2 (Pk2) is a post-synaptic non-canonical Wnt signaling protein shown to interact with post-synaptic density 95 (PSD-95). Here, we show that mice with disruption in Prickle2 display behavioral abnormalities including altered social interaction, learning abnormalities and behavioral inflexibility. Prickle2 disruption in mouse hippocampal neurons led to reductions in dendrite branching, synapse number and PSD size. Consistent with these findings, Prickle2 null neurons show decreased frequency and size of spontaneous miniature synaptic currents. These behavioral and physiological abnormalities in Prickle2 disrupted mice are consistent with ASD-like phenotypes present in other mouse models of ASDs. In 384 individuals with autism, we identified two with distinct, heterozygous, rare, non-synonymous PRICKLE2 variants (p.E8Q and p.V153I) that were shared by their affected siblings and inherited paternally. Unlike wild-type PRICKLE2, the PRICKLE2 variants found in ASD patients exhibit deficits in morphological and electrophysiological assays. These data suggest that these PRICKLE2 variants cause a critical loss of PRICKLE2 function. The data presented here provide new insight into the biological roles of Prickle2, its behavioral importance, and suggest disruptions in non-canonical Wnt genes such as PRICKLE2 may contribute to synaptic abnormalities underlying ASDs.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/genética , Dendritas/ultraestructura , Hipocampo/patología , Hipocampo/fisiopatología , Proteínas con Dominio LIM/deficiencia , Proteínas con Dominio LIM/fisiología , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/fisiología , Potenciales Postsinápticos Miniatura , Mutación Missense , Neuronas/fisiología , Mutación Puntual , Vía de Señalización Wnt , Secuencia de Aminoácidos , Animales , Células Cultivadas , Trastornos Generalizados del Desarrollo Infantil/fisiopatología , Trastornos Generalizados del Desarrollo Infantil/psicología , Condicionamiento Clásico , Conducta Exploratoria , Miedo , Femenino , Reacción Cataléptica de Congelación/fisiología , Humanos , Proteínas con Dominio LIM/genética , Masculino , Aprendizaje por Laberinto , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Potenciales Postsinápticos Miniatura/genética , Neuronas/patología , Fenotipo , Densidad Postsináptica/patología , Proteínas Recombinantes de Fusión/metabolismo , Homología de Secuencia de Aminoácido , Conducta Social
6.
J Pharmacol Exp Ther ; 338(3): 952-63, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21673071

RESUMEN

Thymidylate synthase (TS) is the only de novo source of thymidylate (dTMP) for DNA synthesis and repair. Drugs targeting TS protein are a mainstay in cancer treatment, but off-target effects and toxicity limit their use. Cytosolic thymidine kinase (TK1) and mitochondrial thymidine kinase (TK2) contribute to an alternative dTMP-producing pathway, by salvaging thymidine from the tumor milieu, and may modulate resistance to TS-targeting drugs. Combined down-regulation of these enzymes is an attractive strategy to enhance cancer therapy. We have shown previously that antisense-targeting TS enhanced tumor cell sensitivity to TS-targeting drugs in vitro and in vivo. Because both TS and TKs contribute to increased cellular dTMP, we hypothesized that TKs mediate resistance to the capacity of TS small interfering RNA (siRNA) to sensitize tumor cells to TS-targeting anticancer drugs. We assessed the effects of targeting TK1 or TK2 with siRNA alone and in combination with siRNA targeting TS and/or TS-protein targeting drugs on tumor cell proliferation. Down-regulation of TK with siRNA enhanced the capacity of TS siRNA to sensitize tumor cells to traditional TS protein-targeting drugs [5-fluorodeoxyuridine (5FUdR) and pemetrexed]. The sensitization was greater than that observed in response to any siRNA used alone and was specific to drugs targeting TS. Up-regulation of TK1 in response to combined 5FUdR and TS siRNA suggests that TK knockdown may be therapeutically useful in combination with these agents. TKs may be useful targets for cancer therapy when combined with molecules targeting TS mRNA and TS protein.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Floxuridina/farmacología , Glutamatos/farmacología , Guanina/análogos & derivados , ARN Interferente Pequeño/farmacología , Timidina Quinasa/antagonistas & inhibidores , Timidilato Sintasa/antagonistas & inhibidores , Actinas/metabolismo , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Regulación hacia Abajo/efectos de los fármacos , Guanina/farmacología , Células HeLa , Humanos , Pemetrexed , Transfección
7.
J Med Genet ; 42(7): 551-7, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15994876

RESUMEN

BACKGROUND: Majeed syndrome is an autosomal recessive, autoinflammatory disorder characterised by chronic recurrent multifocal osteomyelitis and congenital dyserythropoietic anaemia. The objectives of this study were to map, identify, and characterise the Majeed syndrome causal gene and to speculate on its function and role in skin and bone inflammation. METHODS: Six individuals with Majeed syndrome from two unrelated families were identified for this study. Homozygosity mapping and parametric linkage analysis were employed for the localisation of the gene responsible for Majeed syndrome. Direct sequencing was utilised for the identification of mutations within the genes contained in the region of linkage. Expression studies and in silico characterisation of the identified causal gene and its protein were carried out. RESULTS: The phenotype of Majeed syndrome includes inflammation of the bone and skin, recurrent fevers, and dyserythropoietic anaemia. The clinical picture of the six affected individuals is briefly reviewed. The gene was mapped to a 5.5 cM interval (1.8 Mb) on chromosome 18p. Examination of genes in this interval led to the identification of homozygous mutations in LPIN2 in affected individuals from the two families. LPIN2 was found to be expressed in almost all tissues. The function of LPIN2 and its role in inflammation remains unknown. CONCLUSIONS: We conclude that homozygous mutations in LPIN2 result in Majeed syndrome. Understanding the aberrant immune response in this condition will shed light on the aetiology of other inflammatory disorders of multifactorial aetiology including isolated chronic recurrent multifocal osteomyelitis, Sweet syndrome, and psoriasis.


Asunto(s)
Anemia Diseritropoyética Congénita/genética , Homocigoto , Mutación , Proteínas Nucleares/genética , Osteomielitis/genética , Adulto , Animales , Causalidad , Enfermedad Crónica , Secuencia Conservada , Análisis Mutacional de ADN , Familia , Femenino , Ligamiento Genético , Humanos , Jordania , Masculino , Especificidad de Órganos/genética , Linaje , Fenotipo , Recurrencia , Síndrome de Sweet/genética , Síndrome
8.
Br J Pharmacol ; 134(7): 1437-46, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11724749

RESUMEN

1. Thymidylate synthase (TS) is a target for several anticancer drugs. We previously showed that an antisense oligodeoxynucleotide (ODN) directed against TS mRNA down-regulated TS protein and enhanced cytotoxicity of TS-targeting drugs [including 5-fluorodeoxyuridine (5-FUdR)] in HeLa cells. Patient tumours with increased TS expression are resistant to TS-targeting drugs. It was hypothesized that TS mRNA and consequently TS protein could be down-regulated in 5-FUdR-resistant cells that overexpress TS, sensitizing them to 5-FUdR cytotoxicity. In this study we assessed the capacity of an anti-TS antisense ODN to circumvent resistance dependent on TS overexpression. 2. Variant HeLa clones exhibiting 2 - 20 fold resistance to 5-FUdR were selected by exposing cultured cells to drug. Clones FUdR-5a, -25b, and -50a expressed TS protein levels 10 fold, 10 fold, and 17 fold higher (respectively) than parental cells. Cells were treated with antisense ODN 83 (a 2'-methoxy-ethoxylated, phosphorothioated 20-mer, complementary to a portion of the 3'-untranslated region of TS mRNA), or ODN 32 (a control ODN with the same base composition as ODN 83, but in randomized order). Twenty-four and 48 h following transfection (50-100 nM ODN, plus polycationic liposome), TS mRNA levels (by RT-PCR) and protein levels (by radiolabelled 5-FUdR-monophosphate binding) were decreased by at least 60% in ODN 83-treated cells compared with control ODN 32-treated cells. ODN 83 enhanced the cytotoxicity of 5-FUdR by up to 85% in both parental and 5-FUdR-resistant cell lines. 3. Antisense ODN can be used to down-regulate TS and attenuate drug resistance in TS-overexpressing cells.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , División Celular/efectos de los fármacos , ADN sin Sentido/farmacología , Floxuridina/farmacología , Timidilato Sintasa/efectos de los fármacos , División Celular/genética , ADN sin Sentido/genética , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Resistencia a Antineoplásicos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Timidilato Sintasa/genética , Timidilato Sintasa/metabolismo , Factores de Tiempo , Transfección
9.
J Pharmacol Exp Ther ; 298(2): 477-84, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11454908

RESUMEN

Chemotherapeutic agents targeting thymidylate synthase (TS) are effective against human tumors. Efficacy is limited by drug resistance, often mediated by TS overexpression. Treatment of HeLa cells in vitro with an antisense oligodeoxynucleotide (ODN 83) targeting human TS mRNA reduces TS mRNA and protein levels, inhibits cell proliferation, and sensitizes cells to TS-targeting drugs (Ferguson et al., 1999). The present study investigates the mechanism by which ODN 83 inhibits cell proliferation and examines its antitumor efficacy in vivo. ODN 83 treatment did not induce apoptosis in HeLa cells in vitro but caused accumulation of cells at G2/M. In contrast, TS-targeting chemotherapeutics arrest at G1 or S. Antisense down-regulation reduced TS mRNA levels in human colon cancer (HT29) cells by 40% in vitro, resulted in G2/M arrest, and reduced proliferation without enhanced cell death. Growth of HT29 tumors in immunocompromised mice was significantly inhibited when antisense ODN 83 treatment began promptly after tumor implantation and was accompanied by a 40% reduction in TS protein levels. Growth of tumors allowed to reach 400 mm3 prior to ODN administration was unaffected by antisense ODN 83. Radiolabeled ODNs were localized to the tumor periphery but evenly distributed in normal tissue. Thus, down-regulation of TS mRNA and protein by antisense ODN treatment exerts a novel G2/M cell cycle block without increasing cell death and inhibits HT29 tumor cell growth in vivo. Antisense ODN 83 may be an effective therapy for colon carcinoma, alone or in combination with TS-targeting cytotoxic drugs.


Asunto(s)
Antineoplásicos/farmacología , Oligonucleótidos Antisentido/farmacología , Timidilato Sintasa/genética , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Femenino , Citometría de Flujo , Células HT29 , Células HeLa , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
10.
Nat Genet ; 27(1): 20-1, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11137993

RESUMEN

IPEX is a fatal disorder characterized by immune dysregulation, polyendocrinopathy, enteropathy and X-linked inheritance (MIM 304930). We present genetic evidence that different mutations of the human gene FOXP3, the ortholog of the gene mutated in scurfy mice (Foxp3), causes IPEX syndrome. Recent linkage analysis studies mapped the gene mutated in IPEX to an interval of 17-20-cM at Xp11. 23-Xq13.3.


Asunto(s)
Proteínas de Unión al ADN/genética , Ligamiento Genético/genética , Mutación/genética , Poliendocrinopatías Autoinmunes/genética , Enteropatías Perdedoras de Proteínas/genética , Cromosoma X/genética , Secuencia de Aminoácidos , Animales , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Femenino , Factores de Transcripción Forkhead , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Linaje , Fenotipo , Alineación de Secuencia , Síndrome
11.
Am J Med Genet ; 90(5): 390-7, 2000 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-10706361

RESUMEN

The clinical findings of a kindred with an X-linked disorder are characterized by autoimmune polyendocrinopathy, enteropathy with villous atrophy, chronic dermatitis, and variable immunodeficiency. Linkage analysis was performed on 20 members of the affected kindred to determine the location of the responsible locus. Informative recombinations limited the region to an approximate 20 cM interval bordered by DXS1055 and DXS1196/DXS1050. Multipoint analysis generated a lod score >3 for the region contained between DXS8024 and DXS8031. The candidate region includes the Wiskott-Aldrich syndrome (WAS) locus. Evaluation of the Wiskott-Aldrich syndrome protein gene by single strand conformational analysis, heteroduplex analysis, and direct sequencing of the 12 exons in an affected male and two carrier females revealed no abnormalities. We conclude that this kindred has an X-linked disorder, distinct from WAS, that results in autoimmunity and variable immunodeficiency. The responsible locus maps to the pericentromeric region Xp11.23 to Xq21.1.


Asunto(s)
Autoinmunidad , Síndromes de Inmunodeficiencia/genética , Proteínas/genética , Aberraciones Cromosómicas Sexuales/diagnóstico , Cromosoma X/genética , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Ligamiento Genético , Humanos , Lactante , Masculino , Linaje , Reacción en Cadena de la Polimerasa , Proteínas/análisis , Síndrome de Wiskott-Aldrich/genética , Proteína del Síndrome de Wiskott-Aldrich
12.
Drug Metab Dispos ; 27(12): 1399-405, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10570020

RESUMEN

Platinum drugs comprise one of the main classes of chemotherapy drugs that can induce remissions in various solid tumors. Although tumors often regress on treatment with cis-diamminedichloroplatinum II (cisplatin) or cis-diammine-1,1-cyclobutane dicarboxylate platinum II (carboplatin), they usually relapse as a drug-resistant tumor. Most mechanisms of platinum resistance could be overcome by increasing the amount of drug that is accumulated by tumor cells. Amphotericin B (Amph B) is efficient at increasing platinum drug uptake, but because of nephrotoxicity associated with extended usage, and the potential for synergistic nephrotoxicity when used with platinum drugs, Amph B has not been used clinically for this purpose. A liposomal preparation of Amph B (LipoAmph B), which is substantially less nephrotoxic, was studied for its ability to enhance platinum-drug toxicity to a human oral squamous cell carcinoma line, HN-5a, and its carboplatin-resistant variant, 5a/carbo-15a, in which cisplatin accumulation was reduced by approximately 40%. Amph B at 10 microg/ml enhanced cisplatin accumulation by approximately 100% in both cell lines, enhancing cytotoxicity of the drugs by 35 to 60%, and completely reversed resistance to both cisplatin and carboplatin. LipoAmph B in the presence of phospholipase A(2)-II (PLA2-II) was able to enhance cisplatin and carboplatin cytotoxicity as effectively as free Amph B in both cell lines. At optimal concentrations, LipoAmph B plus PLA2-II enhanced drug uptake sufficiently to abolish resistance in the platinum-resistant line. Because PLA2-II is elevated in some tumor microenvironments and in plasma of ill patients, LipoAmph B has potential clinical usefulness as a modulator of platinum-drug efficacy.


Asunto(s)
Anfotericina B/farmacología , Antineoplásicos/farmacología , Cisplatino/farmacología , Fosfolipasas A/farmacología , Anfotericina B/uso terapéutico , Antineoplásicos/metabolismo , Antineoplásicos/uso terapéutico , Transporte Biológico , Carboplatino/farmacología , Carcinoma de Células Escamosas , Cisplatino/metabolismo , Cisplatino/uso terapéutico , Resistencia a Antineoplásicos/fisiología , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Fosfolipasas A2 Grupo II , Neoplasias de Cabeza y Cuello , Humanos , Liposomas , Fosfolipasas A2 , Células Tumorales Cultivadas
13.
Br J Pharmacol ; 127(8): 1777-86, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10482907

RESUMEN

1. Thymidylate synthase (TS), the key enzyme in de novo synthesis of thymidine, is an important target for antitumour chemotherapy. It was hypothesized that antisense oligonucleotide down-regulation of TS mRNA would decrease TS levels and enhance the cytotoxicity of inhibitors of TS, including the pyrimidine analogues 5-fluorouracil (5-FU) and 5-fluorodeoxyuridine (5-FUdR), and the folate analogue Tomudex (ICI D1694; N-(5-[N-(3, 4-dihydro-2-methyl-4-oxoquinazolin-6-ylmethyl)-N-methylamino ]-2-theon yl-L-glutamic acid). 2. 2'-Methoxyethoxylated, phosphorothioated 20-mer oligodeoxynucleotides (ODNs), complementary to various sequences in TS mRNA, were synthesized, along with control oligomers consisting of the same, respective bases in randomized order, against which all the biological effects were compared. Following a 6-h transfection of HeLa cells using polycationic liposome at 3 microg ml(-1), ODN 83 (50 nM), complementary to a region in the 3'-untranslated region of the TS mRNA, decreased TS mRNA levels by approximately 70% within 24 h. ODN 83 also decreased TS enzyme activity, as measured by binding of TS to radiolabelled 5-fluorodeoxyuridine monophosphate. In addition to inhibiting proliferation by up to approximately 40%, ODN 83 enhanced the cytotoxicity of Tomudex or 5-FU, added 1 day following transfection, by 50 - 60%. ODN 83 also enhanced sensitivity to 5-FUdR by 70%, but did not affect the toxicity of cisplatin, chlorambucil, melphalan, doxorubicin, ionizing radiation, paclitaxel, or irinotecan. 3. These data indicate that antisense ODN down-regulation of TS can inhibit human tumour cell proliferation and enhance the efficacy of TS-targeted drugs.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Floxuridina/farmacología , Fluorouracilo/farmacología , Oligonucleótidos Antisentido/farmacología , Quinazolinas/farmacología , Tiofenos/farmacología , Timidilato Sintasa/antagonistas & inhibidores , Recuento de Células/efectos de los fármacos , Regulación hacia Abajo , Femenino , Células HeLa , Humanos , Oligonucleótidos Antisentido/genética , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Timidilato Sintasa/genética , Timidilato Sintasa/metabolismo , Transfección
14.
J Pharmacol Exp Ther ; 288(3): 1185-91, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10027857

RESUMEN

Cellular uptake of many chemotherapeutic nucleoside analogs is dependent on the activity of a family of nucleoside transport proteins located in the cell plasma membrane. In the present study, we examined the role of these transporters in the accumulation of gemcitabine by a human head and neck squamous carcinoma cell line. The uptake of [3H]gemcitibine was compared with that of [3H]uridine and [3H]formycin B in the parent cell line (HN-5a) and in a gemcitabine-resistant variant (GEM-8e). The HN-5a and GEM-8e cells were similar in their transport characteristics and expressed predominantly the es (equilibrative, inhibitor-sensitive) transporter subtype; less than 10% of the influx of [3H]formycin B or [3H]uridine was mediated by the ei (equilibrative inhibitor-resistant) system, and there was no evidence for Na+-dependent nucleoside transporters. [3H]Gemcitabine (10 microM) entered these cells via both the es and ei transporters with an initial rate of uptake similar to that seen with the use of [3H]formycin B or [3H]uridine. In addition, ATP-replete cells accumulated significantly less [3H]gemcitabine than did ATP-depleted cells, which is indicative of an active efflux mechanism for gemcitabine. These results show that gemcitabine is a substrate for both the es and ei nucleoside transporters of HN-5a and GEM-8e cells and that gemcitabine resistance of the GEM-8e cells cannot be attributed to changes in transporter activity. Further studies to define the characteristics of the putative efflux mechanism are clearly warranted because this system has the potential to significantly affect the clinical efficacy of gemcitabine.


Asunto(s)
Antimetabolitos Antineoplásicos/metabolismo , Carcinoma de Células Escamosas/metabolismo , Proteínas Portadoras/metabolismo , Desoxicitidina/análogos & derivados , Neoplasias de Cabeza y Cuello/metabolismo , Proteínas de la Membrana/metabolismo , Adenosina Trifosfato/metabolismo , Desoxicitidina/metabolismo , Resistencia a Medicamentos , Formicinas/metabolismo , Humanos , Proteínas de Transporte de Nucleósidos , Factores de Tiempo , Tritio , Células Tumorales Cultivadas , Uridina/metabolismo , Gemcitabina
15.
J Pharmacol Exp Ther ; 286(3): 1333-40, 1998 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9732397

RESUMEN

The uptake of [3H]formycin B by Ehrlich ascites tumor cells was examined in both normal Na+ buffer (physiological) and nominally Na+-free buffer (iso-osmotic replacement with Li+). These studies were conducted to further characterize the equilibrative nucleoside transporter subtypes of Ehrlich cells and to assess the contribution of Na+-dependent concentrative transport mechanisms to the cellular accumulation of nucleoside analogues by these cells. Formycin B is poorly metabolized by mammalian cells and, hence, can be used as a substrate to measure transport kinetics in energetically competent cells. Initial studies established that formycin B inhibited [3H]uridine uptake by the ei (equilibrative inhibitor-insensitive) and es (equilibrative inhibitor-sensitive) transporters of Ehrlich cells with Ki values of 48 +/- 28 and 277 +/- 25 microM, respectively. Similarly, [3H]formycin B had Km values of 111 +/- 52 and 635 +/- 147 microM for uptake by the ei and es transporters, respectively. When assays were conducted in the presence of Na+, plus 100 nM nitrobenzylthioinosine to prevent efflux via the es transporters, the intracellular concentration of [3H]formycin B exceeded the initial medium concentration by more than 3-fold, indicating the activity of a Na+-dependent transporter. Interestingly, the initial rate of uptake of [3H]formycin B was significantly higher in the Li+ buffer (es-mediated Vmax = 65 +/- 10 pmol/microliter . sec) than in the Na+ buffer (Vmax = 8.4 +/- 0.9 pmol/microliter . sec); this may reflect trans-acceleration of [3H]formycin B uptake by elevated intracellular adenosine levels resulting from the low Na+ environment. This model was then used to assess the interaction of gemcitabine (2',2'-difluorodeoxycytidine) with the equilibrative and concentrative nucleoside transporters. Gemcitabine, which has shown considerable potential for the treatment of solid tumors, was a relatively poor inhibitor of [3H]formycin B uptake via the equilibrative transporters (IC50 approximately 400 microM). In contrast, gemcitabine was a potent inhibitor of the Na+-dependent nucleoside transporter of Ehrlich cells (IC50 = 17 +/- 5 nM). These results suggest that the cellular expression/activity of Na+-dependent nucleoside transporters may be an important determinant in gemcitabine cytotoxicity and clinical efficacy.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Proteínas Portadoras/metabolismo , Desoxicitidina/análogos & derivados , Formicinas/metabolismo , Proteínas de la Membrana/metabolismo , Sodio/farmacología , Animales , Carcinoma de Ehrlich/metabolismo , Desoxicitidina/farmacología , Masculino , Ratones , Proteínas de Transporte de Nucleósidos , Tioinosina/análogos & derivados , Tioinosina/metabolismo , Uridina/metabolismo , Gemcitabina
16.
Blood ; 89(3): 1068-77, 1997 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-9028340

RESUMEN

The primate erythrocyte (E) complement receptor, CR1, is a transmembrane glycoprotein located in clusters on the surface of E. In vivo studies have demonstrated that during processing and clearance of complement-opsonized immune complexes, large amounts of immunoglobulin G (IgG) can be bound to primate E via CR1 with no E loss or lysis. However, when comparable amounts of IgG are bound to other sites on E, in many cases the E are cleared from the circulation by the mononuclear phagocytic system. Therefore, due to its role in immune complex processing, CR1 may represent a privileged site on the primate E. To delineate further this property of E CR1, we performed in vitro phagocytosis assays in the absence of complement and examined the ingestion of E, opsonized at various sites with IgG, by peripheral blood monocytes. When either human or rhesus monkey E were opsonized at sites other than CR1, with between 1,000 and 15,000 IgG per E, substantial phagocytosis of E was evident. However, when comparable amounts of IgG were bound exclusively via CR1, little, if any, phagocytosis was observed. The key to the low phagocytic level of E opsonized via CR1 may be related to the requirements of a "zipper mechanism" for phagocytosis first annunciated by Griffin et al. Based on their findings, we suggest that due to the presence of preexisting clusters of CR1 on the E membrane, large amounts of IgG can be bound to E under conditions that preclude circumferential engagement (and phagocytosis) of the entire E by Fc receptors on the monocyte.


Asunto(s)
Eritrocitos/metabolismo , Inmunoglobulina G/metabolismo , Fagocitosis/inmunología , Receptores de Complemento 3b/sangre , Animales , Sitios de Unión/inmunología , Eritrocitos/inmunología , Monocitos/inmunología , Monocitos/metabolismo , Conejos , Receptores de Complemento 3b/inmunología , Ovinos , Especificidad de la Especie
17.
Clin Immunol Immunopathol ; 82(1): 49-59, 1997 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9000042

RESUMEN

In the circulation of primates, C3b-opsonized immune complexes (IC) bound to erythrocyte (E) CR1 are taken to the liver and spleen where IC are removed and destroyed without lysis or sequestration of E. Individuals with diseases associated with IC processing often have decreased E CR1 levels, and in previous primate animal models of IC disease, E CR1 was shown to be reduced, but the relationship between IC processing and CR1 loss remained to be clarified. We have developed a simple model to study this question. In naive (nonimmunized) rhesus monkeys, E-bound mouse anti-CR1 mAbs (1500 IgG/E) are not rapidly cleared from the circulation. Infusion of monkey anti-mouse IgG leads to rapid indirect binding of this second antibody to E CR1. Subsequently, in what appears to be a concerted reaction, CR1-bound nascent IC are rapidly cleared from the circulation and CR1 is removed from E at the same rate. Clearance of bound IC and loss of CR1 were both independently followed by RIA. Imaging studies localized the cleared anti-CR1 mAbs to the liver. Western blots indicated that the loss of CR1 was not due to a conformational change, and E CR1 levels returned to normal in 2-3 weeks, suggesting that the return was associated with synthesis of new E. Our findings suggest that the key step in the clearance mechanism requires recognition (possibly by Fc receptors) of IC-like material associated with E CR1, and this leads to loss of CR1 and uptake of the CR1-IC substrate by liver phagocytic cells.


Asunto(s)
Complejo Antígeno-Anticuerpo/metabolismo , Eritrocitos/química , Macaca mulatta/sangre , Receptores de Complemento/inmunología , Receptores de Complemento/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Western Blotting , Complemento C3b/metabolismo , Epítopos/análisis , Eritrocitos/inmunología , Radioisótopos de Yodo , Hígado/metabolismo , Ratones , Modelos Inmunológicos , Radioinmunoensayo
18.
J Immunol ; 158(2): 842-50, 1997 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-8993002

RESUMEN

We investigated the feasibility of using bispecific mAb complexes to redirect and improve the efficiency of the primate E complement receptor 1-based clearance reaction to remove a virus from the circulation. As an initial approach, we used bacteriophage phiX174 as an immunologic model for mammalian viruses. Bispecific complexes were prepared by chemically cross-linking a mAb specific for complement receptor 1 with a mAb specific for the bacteriophage phiX174. In a monkey model these complexes facilitate rapid and quantitative binding of the target bacteriophage to E in vitro and in vivo. Moreover, after in vivo binding to E, the complexes containing mAb and prototype virus are rapidly cleared from the circulation of rhesus and cynomolgus monkeys without loss of E. Our findings suggest that bispecific mAb complexes, in concert with primate E complement receptor 1, may have therapeutic utility in the treatment of diseases associated with blood-borne pathogens.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/metabolismo , Complejo Antígeno-Anticuerpo/inmunología , Bacteriófago phi X 174/inmunología , Eritrocitos/inmunología , Receptores de Complemento/inmunología , Receptores de Complemento/metabolismo , Virus/inmunología , Virus/metabolismo , Animales , Modelos Animales de Enfermedad , Eritrocitos/virología , Macaca mulatta , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...