Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 169
Filtrar
1.
AJNR Am J Neuroradiol ; 38(2): 376-382, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27765741

RESUMEN

BACKGROUND AND PURPOSE: Infantile neuronal ceroid lipofuscinosis is a devastating neurodegenerative storage disease caused by palmitoyl-protein thioesterase 1 deficiency, which impairs degradation of palmitoylated proteins (constituents of ceroid) by lysosomal hydrolases. Consequent lysosomal ceroid accumulation leads to neuronal injury, resulting in rapid neurodegeneration and childhood death. As part of a project studying the treatment benefits of a combination of cysteamine bitartrate and N-acetyl cysteine, we made serial measurements of patients' brain volumes with MR imaging. MATERIALS AND METHODS: Ten patients with infantile neuronal ceroid lipofuscinosis participating in a treatment/follow-up study underwent brain MR imaging that included high-resolution T1-weighted images. After manual placement of a mask delineating the surface of the brain, a maximum-likelihood classifier was applied to determine total brain volume, further subdivided as cerebrum, cerebellum, brain stem, and thalamus. Patients' brain volumes were compared with those of a healthy population. RESULTS: Major subdivisions of the brain followed similar trajectories with different timing. The cerebrum demonstrated early, rapid volume loss and may never have been normal postnatally. The thalamus dropped out of the normal range around 6 months of age; the cerebellum, around 2 years of age; and the brain stem, around 3 years of age. CONCLUSIONS: Rapid cerebral volume loss was expected on the basis of previous qualitative reports. Because our study did not include a nontreatment arm and because progression of brain volumes in infantile neuronal ceroid lipofuscinosis has not been previously quantified, we could not determine whether our intervention had a beneficial effect on brain volumes. However, the level of quantitative detail in this study allows it to serve as a reference for evaluation of future therapeutic interventions.


Asunto(s)
Encéfalo/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos , Lipofuscinosis Ceroideas Neuronales/diagnóstico por imagen , Acetilcisteína/uso terapéutico , Envejecimiento/patología , Tronco Encefálico/diagnóstico por imagen , Cerebelo/diagnóstico por imagen , Cerebro/diagnóstico por imagen , Niño , Preescolar , Cisteamina/uso terapéutico , Electroencefalografía , Femenino , Estudios de Seguimiento , Humanos , Lactante , Masculino , Lipofuscinosis Ceroideas Neuronales/tratamiento farmacológico , Tálamo/diagnóstico por imagen
2.
J Perinatol ; 33(11): 847-50, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23722974

RESUMEN

OBJECTIVE: Earlier diagnosis and treatment of necrotizing enterocolitis (NEC) in preterm infants, before clinical deterioration, might improve outcomes. A monitor that measures abnormal heart rate characteristics (HRC) of decreased variability and transient decelerations was developed as an early warning system for sepsis. As NEC shares pathophysiologic features with sepsis, we tested the hypothesis that abnormal HRC occur before clinical diagnosis of NEC. STUDY DESIGN: Retrospective review of Bells stage II to III NEC cases among infants <34 weeks gestation enrolled in a prospective randomized clinical trial of HRC monitoring at three neonatal intensive care units. RESULT: Of 97 infants with NEC and HRC data, 33 underwent surgical intervention within 1 week of diagnosis. The baseline HRC index from 1 to 3 days before diagnosis was higher in patients who developed surgical vs medical NEC (2.06±1.98 vs 1.22±1.10, P=0.009). The HRC index increased significantly 16 h before the clinical diagnosis of surgical NEC and 6 h before medical NEC. At the time of clinical diagnosis, the HRC index was higher in patients with surgical vs medical NEC (3.3±2.2 vs 1.9±1.7, P<0.001). CONCLUSION: Abnormal HRC occur before clinical diagnosis of NEC, suggesting that continuous HRC monitoring may facilitate earlier detection and treatment.


Asunto(s)
Enterocolitis Necrotizante/diagnóstico , Enterocolitis Necrotizante/fisiopatología , Frecuencia Cardíaca , Enterocolitis Necrotizante/terapia , Monitoreo del Ambiente , Femenino , Humanos , Recién Nacido , Enfermedades del Prematuro/diagnóstico , Enfermedades del Prematuro/fisiopatología , Masculino , Estudios Prospectivos , Estudios Retrospectivos
4.
DNA Cell Biol ; 20(3): 149-57, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11313018

RESUMEN

The pancreatic secretory phospholipase A(2) (sPLA(2)IB) is considered to be a digestive enzyme, although it has several important receptor-mediated functions. In this study, using the newly isolated murine sPLA(2)IB cDNA clone as a probe, we demonstrate that in addition to the pancreas, the sPLA(2)IB mRNA was expressed in extrapancreatic organs such as the liver, spleen, duodenum, colon, and lungs. We also demonstrate that sPLA(2)IB mRNA expression was detectable from the 17(th) day of gestation in the developing mouse fetus, coinciding with the time of completion of differentiation of the pancreas. Furthermore, the mRNA expression pattern of sPLA(2)IB was distinct from those of sPLA(2)IIA and cPLA(2) in various tissues examined. The murine sPLA(2)IB gene structure is well conserved, consistent with findings in other mammalian species, and this gene mapped to the region of mouse chromosome 5F1-G1.1. Taken together, our results suggest that sPLA(2)IB plays important roles both in the pancreas and in extrapancreatic tissues and that in the mouse, its expression is developmentally regulated.


Asunto(s)
Páncreas/enzimología , Fosfolipasas A/genética , Fosfolipasas A/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Mapeo Cromosómico , Cromosomas Humanos Par 12 , Secuencia Conservada , Ciclooxigenasa 2 , Cisteína , Embrión de Mamíferos/enzimología , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Proteínas de la Membrana , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Prostaglandina-Endoperóxido Sintasas/genética , Prostaglandina-Endoperóxido Sintasas/metabolismo
5.
Nat Med ; 7(4): 478-84, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11283676

RESUMEN

Neuronal ceroid lipofuscinoses (NCLs) are the most common hereditary neurodegenerative diseases of childhood. The infantile form, INCL, is caused by lysosomal palmitoyl-protein thioesterase (PPT) deficiency, which impairs the cleavage of thioester linkages in palmitoylated proteins, preventing their hydrolysis by lysosomal proteinases. Consequent accumulation of these lipid-modified proteins (constituents of ceroid) in lysosomes leads to INCL. Because thioester linkages are susceptible to nucleophilic attack, drugs with this property may have therapeutic potential for INCL. We report here that two such drugs, phosphocysteamine and N-acetylcysteine, disrupt thioester linkages in a model thioester compound, [14C]palmitoyl approximately CoA. Most importantly, in lymphoblasts derived from INCL patients, phosphocysteamine, a known lysosomotrophic drug, mediates the depletion of lysosomal ceroids, prevents their re-accumulation and inhibits apoptosis. Our results define a novel pharmacological approach to lysosomal ceroid depletion and raise the possibility that nucleophilic drugs such as phosphocysteamine hold therapeutic potential for INCL.


Asunto(s)
Ceroide/metabolismo , Lipofuscinosis Ceroideas Neuronales/tratamiento farmacológico , Lipofuscinosis Ceroideas Neuronales/metabolismo , Acetilcisteína/farmacología , Apoptosis/efectos de los fármacos , Células Cultivadas , Niño , Codón sin Sentido , Cistafos/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Glicoproteínas/metabolismo , Humanos , Lactante , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Linfocitos/patología , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Mutación Missense , Lipofuscinosis Ceroideas Neuronales/patología , Palmitoil Coenzima A/metabolismo , Palmitoil-CoA Hidrolasa/deficiencia , Palmitoil-CoA Hidrolasa/genética , Saposinas
6.
Mol Genet Metab ; 70(4): 301-9, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10993717

RESUMEN

Farber disease is an autosomal recessive disorder caused by lysosomal acid ceramidase (AC) deficiency. It commonly manifests during the first few months after birth with a unique triad of painful and progressive deformed joints, subcutaneous nodules, and progressive hoarseness. In order to understand the molecular mechanism(s) of pathogenesis of Farber disease, we isolated and characterized a full-length human AC gene, mapped its chromosomal location, determined the tissue-specific expression, and analyzed mutations in Farber disease patients. We also studied the AC-mRNA expression in gastrointestinal tumors and adjoining normal tissues. In addition, we determined the pattern of tissue-specific AC-mRNA expression in the adult mouse and during fetal development. Our results show that human AC gene consists of 14 exons and 13 introns spanning approximately 26.5 kb of genomic DNA. It is mapped to human chromosome 8p22-21.2, a region often disrupted in several cancers. The AC-mRNA is expressed in the mouse fetus from the seventh day of gestation. Interestingly, while the AC-mRNA is expressed in all segments of the normal gastrointestinal tract, none of the gastrointestinal tumor tissues had any AC-mRNA expression. We also uncovered four novel mutations in Farber disease patients that were not previously reported. Taken together, our results not only attest to the physiological importance of AC but also uncover several new mutations in Farber disease that may advance our knowledge towards establishing a genotype-phenotype correlation in this disease.


Asunto(s)
Amidohidrolasas/genética , Enfermedades por Almacenamiento Lisosomal/genética , Mutación , Ceramidasa Ácida , Animales , Secuencia de Bases , Células Cultivadas , Ceramidasas , Mapeo Cromosómico , Cartilla de ADN , Desarrollo Embrionario y Fetal , Humanos , Hibridación Fluorescente in Situ , Enfermedades por Almacenamiento Lisosomal/enzimología , Ratones , ARN Mensajero/genética
7.
Cancer Res ; 60(4): 1084-91, 2000 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-10706128

RESUMEN

Cyclooxygenase-2 (COX-2) gene overexpression is suggested to play important roles in colorectal tumorigenesis. Epidemiological studies revealed that nonsteroidal anti-inflammatory drugs (NSAIDs), such as aspirin and sulindac, which inhibit COX activity, reduce colorectal cancer mortality. Current investigations have focused on delineating the molecular mechanisms that regulate COX-2 gene expression and the roles of NSAIDs in cancer chemoprevention. COX-2 catalyzes the production of prostaglandins (PGs) from arachidonic acid (AA), generated by phospholipases A2 (PLA2s), a family of acyl esterases that cause the release of AA from cellular phospholipids. Pancreatic secretory PLA2 (sPLA2), via its receptor (sPLA2R), transcriptionally activates COX-2 gene expression in several cell types, although a specific transcription factor mediating COX-2 expression has not yet been identified. Here, we report that a transcription factor, CCAAT/enhancer-binding protein beta(C/EBPbeta), plays a critical role in sPLA2IB-induced, receptor-mediated COX-2 gene expression in MC3T3E1 and NIH3T3 cells. Furthermore, treatment of these cells with NSAIDs in the presence of sPLA2IB appears to potentiate the stimulatory effects on COX-2 mRNA and COX-2 protein expression and a concomitant elevation in PG production. Most significantly, NSAID treatment appears to drastically suppress the production of cytosolic PLA2 (cPLA2) mRNA. The lack of sPLA2IB, sPLA2IIA, and sPLA2V mRNA expression in both NIH3T3 and MC3T3E1 cells suggests that cPLA2 is the most likely enzyme that catalyzes the release of AA, the rate-limiting substrate of COX for the production of PGs. Our results suggest that: (a) sPLA2IB receptor-mediated COX-2 expression is mediated via C/EBPbeta; (b) NSAIDs in the presence of sPLA2IB potentiate the stimulatory effects of sPLA2IB on COX-2 mRNA expression; and (c) despite the apparent stimulation of COX-2 expression by NSAIDs, they strikingly deprive COX-2 of its substrate, AA, by suppressing cPLA2 mRNA expression. Both AA and PGs regulate many vital biological functions (e.g., motility and invasiveness) that are dysregulated in most cancer cells, and they have profound effects on cellular differentiation. Our results raise the possibility that deprivation of COX-2 of its substrate by the suppression of cPLA2 mRNA expression is an additional mechanism used by NSAIDs to inhibit tumorigenesis.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Isoenzimas/genética , Prostaglandina-Endoperóxido Sintasas/genética , Células 3T3 , Animales , Proteínas Potenciadoras de Unión a CCAAT , Ciclooxigenasa 2 , Proteínas de Unión al ADN/fisiología , Ratones , Proteínas Nucleares/fisiología , Fosfolipasas A/genética , Fosfolipasas A/farmacología , Fosfolipasas A2 , Regiones Promotoras Genéticas , Transcripción Genética
9.
Biotechnol Prog ; 16(1): 17-25, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10662484

RESUMEN

Numerous biopharmaceuticals and other recombinant biotechnology products are made in prokaryotic hosts. However, bacterial production of native, biologically active eukaryotic proteins is rarely possible for disulfide-bonded and/or multisubunit proteins. We previously described the production of soluble, native disulfide-bonded dimeric proteins in the Escherichia coli cytoplasm (Miele et al., 1990; Mantile et al., 1993). Native, biologically active proteins with up to six disulfide bonds have been produced with our expression system (Garces et al., 1997). However, plasmid instability during induction limited its usefulness. We now report the stable, high-level expression of soluble, disulfide-bonded human uteroglobin without antibiotic selection. We designed a new vector containing a multifunctional stabilization region that confers complete plasmid stability and increased protein yields without copy number increases. Recombinant expression remains fully inducible after long-term continuous culture in nonselective liquid medium (at least 260 generations). This system may significantly expand the applications of bacterial expression to recombinant production of soluble, bioactive proteins for biochemical studies and biopharmaceutical/industrial purposes. As a result of the very broad activity spectrum of the stabilization region we selected, its use could be extended to bacterial hosts other than enterobacteria.


Asunto(s)
Biofarmacia , Proteínas Recombinantes/biosíntesis , Antibacterianos , Secuencia de Bases , Biotecnología , Dimerización , Disulfuros/química , Escherichia coli/genética , Expresión Génica , Vectores Genéticos , Humanos , Sondas de Oligonucleótidos/genética , Plásmidos/genética , Ingeniería de Proteínas , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Solubilidad , Uteroglobina/biosíntesis , Uteroglobina/química , Uteroglobina/genética
10.
J Biol Chem ; 275(2): 969-76, 2000 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-10625634

RESUMEN

Osteopontin (OPN) is a sialic acid-rich, adhesive, extracellular matrix (ECM) protein with Arg-Gly-Asp cell-binding sequence that interacts with several integrins, including alpha(v)beta(3). Since the ECM is a key regulator of mammary gland morphogenesis, and mammary epithelial cells express OPN at elevated levels, we sought to determine whether this protein plays a role in the postnatal mammary gland development. By generating transgenic mice that express OPN antisense-RNA (AS-OPN mice) in the mammary epithelia we achieved suppression of OPN production in this organ. The pregnant AS-OPN mice displayed a lack of mammary alveolar structures, a drastic reduction in the synthesis of beta-casein, whey acidic milk protein, and lactation deficiency. In agreement with these findings, we uncovered that a mammary cell line, NMuMG, which undergoes both structural and functional differentiation on ECM-coated plates, when transfected with an antisense OPN-cDNA construct, failed to undergo such differentiation. Furthermore, the results of gel-invasion assays demonstrated that these cells manifest elevated matrix metalloproteinase (MMP) activity when OPN expression is significantly reduced. The identity of this proteinase as MMP-2 is confirmed by Western blotting, zymography, and inhibition of its activity by a specific inhibitor, TIMP-2. Taken together, our results demonstrate, for the first time, an essential role of OPN in mammary gland differentiation and that the molecular mechanism(s) of its action, at least in part, involves down-regulation of MMP-2.


Asunto(s)
Lactancia/genética , Glándulas Mamarias Animales/anomalías , Glándulas Mamarias Animales/fisiología , ARN sin Sentido/genética , Sialoglicoproteínas/genética , Animales , Animales Recién Nacidos , Caseínas/genética , Citocinas/genética , Muerte , Células Epiteliales/fisiología , Femenino , Regulación de la Expresión Génica , Silenciador del Gen , Glándulas Mamarias Animales/embriología , Ratones , Ratones Transgénicos , Proteínas de la Leche/genética , Morfogénesis , Osteopontina , Embarazo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sialoglicoproteínas/deficiencia
11.
Ann N Y Acad Sci ; 923: 113-27, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11193750

RESUMEN

Uteroglobin, a steroid-inducible, cytokine-like, secreted protein with immunomodulatory properties, has been reported to bind progesterone, polychlorinated biphenyls (PCB), and retinol. Structural studies may delineate whether binding of ligands is a likely physiological function of human uteroglobin (hUG). We report a refined crystal structure of uncomplexed recombinant hUG (rhUG) at 2.5-A resolution and the results of our molecular modeling studies of ligand binding to the central hydrophobic cavity of rhUG. The crystal structure of rhUG is very similar to that of reported crystal structures of uteroglobins. Using molecular modeling techniques, the three ligands--PCB, progesterone, and retinol--were docked into the hydrophobic cavity of the dimer structure of rhUG. We undocked the progesterone ligand by pulling the ligand from the cavity into the solvent. From our modeling and undocking studies of progesterone, it is clear that these types of hydrophobic ligands could slip into the cavity between helix-3 and helix-3' of the dimer instead of between helix-1 and helix-4 of the monomer, as proposed earlier. Our results suggest that at least one of the physiological functions of UG is to bind to hydrophobic ligands, such as progesterone and retinol.


Asunto(s)
Modelos Moleculares , Unión Proteica/fisiología , Estructura Terciaria de Proteína/fisiología , Proteínas Recombinantes/química , Uteroglobina/química , Animales , Cristalografía por Rayos X/métodos , Humanos , Ligandos
12.
Ann N Y Acad Sci ; 923: 210-33, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11193759

RESUMEN

To determine the physiological function(s) of uteroglobin (UG), a steroid-inducible, homodimeric, secreted protein, we have generated transgenic mice that either are completely UG-deficient due to UG gene-knockout (UG-KO) or are partially UG-deficient due to the expression of UG antisense RNA (UG-AS). Both the UG-KO and UG-AS mice develop immunoglobulin A (IgA) nephropathy (IgAN), characterized by microhematuria, albuminuria, and renal glomerular deposition of IgA, fibronectin (Fn), collagen, and C3 complement. This phenotype of both UG-KO and UG-AS mice is virtually identical to that of human IgAN, the most common primary glomerulopathy worldwide. The molecular mechanism by which UG prevents this disease in mice appears to center around UG's interaction with Fn. Since Fn, IgA, and UG are present in circulation and high plasma levels of IgA-Fn complex have been reported in human IgAN, we sought to determine whether UG interacts with Fn and prevents Fn-Fn and/or IgA-Fn interactions, essential for abnormal tissue deposition of Fn and IgA. Our coimmunoprecipitation studies uncovered the formation of Fn-UG heteromers in vitro and these heteromers are detectable in the plasma of normal mice, but not UG-KO mice. Further, high plasma levels of IgA-Fn complex, a characteristic of human IgAN patients, were also found in UG-KO mice. Finally, coadministration of UG + Fn or UG + IgA to UG-KO mice prevented glomerular deposition of Fn and IgA, respectively. Our results define a possible molecular mechanism of IgAN and provide insight into at least one important physiological function of UG in maintaining normal renal function in mice.


Asunto(s)
Glomerulonefritis por IGA/genética , Riñón/metabolismo , Ratones Noqueados/metabolismo , Ratones Transgénicos/metabolismo , ARN sin Sentido , Uteroglobina/deficiencia , Uteroglobina/genética , Animales , Colágeno/genética , Complemento C3/metabolismo , Fibronectinas/sangre , Fibronectinas/genética , Mesangio Glomerular/inmunología , Mesangio Glomerular/metabolismo , Mesangio Glomerular/patología , Glomerulonefritis por IGA/inmunología , Glomerulonefritis por IGA/patología , Hematuria/inmunología , Hematuria/patología , Hematuria/fisiopatología , Humanos , Inmunoglobulina A/sangre , Riñón/patología , Riñón/fisiopatología , Ratones , Ratones Noqueados/genética , Ratones Noqueados/inmunología , Ratones Transgénicos/genética , Ratones Transgénicos/inmunología , Fenotipo , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , ARN sin Sentido/genética , ARN Mensajero/metabolismo , Uteroglobina/biosíntesis
13.
Ann N Y Acad Sci ; 923: 234-48, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11193760

RESUMEN

Uteroglobin (UG) is a multifunctional, secreted protein with anti-inflammatory and antichemotactic properties. While its anti-inflammatory effects, in part, stem from the inhibition of soluble phospholipase A2 (sPLA2) activity, the mechanism(s) of its antichemotactic effects is not clearly understood. Although specific binding of UG on microsomal and plasma membranes has been reported recently, how this binding affects cellular function is not clear. Here, we report that recombinant human UG (hUG) binds to both normal and cancer cells with high affinity (20-35 nM, respectively) and specificity. Affinity cross-linking studies revealed that 125I-hUG binds to the NIH 3T3 cell surface with two proteins of apparent molecular masses of 190 and 49 kDa, respectively. UG affinity chromatography yielded similar results. While both the 190- and 49-kDa proteins were expressed in the heart, liver, and spleen, the lung and trachea expressed only the 190-kDa protein. Some cancer cells (e.g., mastocytoma, sarcoma, and lymphoma) expressed both the 190- and 49-kDa proteins. Further, using functional assays, we found that UG dramatically suppressed the motility and extracellular matrix invasion of both NIH 3T3 and some cancer cells. In order to further characterize the anti-ECM-invasive properties of UG, we induced expression of hUG into cancer cell lines derived from organs that, under physiological circumstances, secrete UG at a high level. Interestingly, it has been reported that a high percentage of the adenocarcinomas arising from the same organs fail to express UG. Our results on induced hUG expression in these cells show that inhibition of motility and ECM invasion requires the expression of both UG and its binding proteins. Taken together, our data define receptor-mediated functions of UG in which this protein regulates vital cellular functions by both autocrine and paracrine pathways.


Asunto(s)
Proteínas Portadoras/metabolismo , Movimiento Celular/genética , Invasividad Neoplásica/genética , Unión Proteica/genética , Receptores de Superficie Celular/metabolismo , Células Tumorales Cultivadas/metabolismo , Uteroglobina/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/farmacología , Movimiento Celular/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Humanos , Ratones , Invasividad Neoplásica/patología , Unión Proteica/efectos de los fármacos , Ensayo de Unión Radioligante/estadística & datos numéricos , Receptores de Superficie Celular/efectos de los fármacos , Receptores de Superficie Celular/aislamiento & purificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/patología , Uteroglobina/genética , Uteroglobina/farmacología
17.
Nat Med ; 5(9): 1018-25, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10470078

RESUMEN

The molecular mechanism(s) of immunoglobulin A (IgA) nephropathy, the most common primary renal glomerular disease worldwide, is unknown. Its pathologic features include hematuria, high levels of circulating IgA-fibronectin (Fn) complexes, and glomerular deposition of IgA, complement C3, Fn and collagen. We report here that two independent mouse models (gene knockout and antisense transgenic), both manifesting deficiency of an anti-inflammatory protein, uteroglobin (UG), develop almost all of the pathologic features of human IgA nephropathy. We further demonstrate that Fn-UG heteromerization, reported to prevent abnormal glomerular deposition of Fn and collagen, also abrogates both the formation of IgA-Fn complexes and their binding to glomerular cells. Moreover, UG prevents glomerular accumulation of exogenous IgA in UG-null mice. These results define an essential role for UG in preventing mouse IgA nephropathy and warrant further studies to determine if a similar mechanism(s) underlies the human disease.


Asunto(s)
Glomerulonefritis por IGA/patología , Inmunoglobulina A/inmunología , Glomérulos Renales/patología , Uteroglobina/fisiología , Animales , Complejo Antígeno-Anticuerpo/análisis , Complejo Antígeno-Anticuerpo/sangre , Complejo Antígeno-Anticuerpo/efectos de los fármacos , Complejo Antígeno-Anticuerpo/inmunología , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Complemento C3/análisis , Complemento C3/inmunología , Modelos Animales de Enfermedad , Fibronectinas/análisis , Fibronectinas/sangre , Fibronectinas/genética , Fibronectinas/inmunología , Eliminación de Gen , Mesangio Glomerular/citología , Mesangio Glomerular/efectos de los fármacos , Mesangio Glomerular/inmunología , Mesangio Glomerular/patología , Glomerulonefritis por IGA/genética , Glomerulonefritis por IGA/inmunología , Glomerulonefritis por IGA/fisiopatología , Hematuria/patología , Hematuria/orina , Humanos , Inmunoglobulina A/análisis , Inmunoglobulina A/sangre , Inmunoglobulina A/efectos de los fármacos , Glomérulos Renales/inmunología , Glomérulos Renales/metabolismo , Glomérulos Renales/fisiopatología , Ratones , Ratones Noqueados , Ratones Transgénicos , Factor de Crecimiento Derivado de Plaquetas/análisis , Factor de Crecimiento Derivado de Plaquetas/genética , Proteínas Proto-Oncogénicas/análisis , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-sis , ARN sin Sentido/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Uteroglobina/deficiencia , Uteroglobina/genética , Uteroglobina/farmacología
18.
Cell Mol Life Sci ; 55(5): 771-87, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10379362

RESUMEN

Blastokinin or uteroglobin (UG) is a steroid-inducible, evolutionarily conserved, multifunctional protein secreted by the mucosal epithelial of virtually all mammals. It is present in the blood and in other body fluids including urine. An antigen immunoreactive to UG antibody is also detectable in the mucosal epithelia of all vertebrates. UG-binding proteins (putative receptor), expressed on several normal and cancer cell types, have been characterized. The human UG gene is mapped to chromosome 11q12.2 13.1, a region that is frequently rearranged or deleted in many cancers. The generation of UG knockout mice revealed that disruption of this gene causes: (i) severe renal disease due to an abnormal deposition of fibronectin and collagen in the glomeruli; (ii) predisposition to a high incidence of malignancies; and (iii) a lack of polychlorinated biphenyl binding and increased oxygen toxicity in the lungs. The mechanism(s) of UG action is likely to be even more complex as it also functions via a putative receptor-mediated pathway that has not yet been clearly defined. Molecular characterization of the UG receptor and signal transduction via this receptor pathway may show that this protein belongs to a novel cytokine/chemokine family.


Asunto(s)
Citocinas/fisiología , Uteroglobina/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , Citocinas/química , Citocinas/genética , ADN Complementario/genética , Regulación de la Expresión Génica , Genes Supresores de Tumor , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Receptores de Citocinas/fisiología , Homología de Secuencia de Aminoácido , Uteroglobina/química , Uteroglobina/genética
19.
Gene ; 231(1-2): 203-11, 1999 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-10231585

RESUMEN

Mutations in the palmitoyl-protein thioesterase (PPT) gene cause infantile neuronal ceroid lipofuscinosis (INCL), the clinical manifestations of which include the early loss of vision followed by deterioration of brain functions. To gain insight into the temporal onset of these clinical manifestations, we isolated and characterized a murine PPT (mPPT)-cDNA, mapped the gene on distal chromosome 4, and studied its expression in the eye and in the brain during development. Our results show that both cDNA and protein sequences of the murine and human PPTs are virtually identical and that the mPPT expression in the retina and in the brain is temporally regulated during development. Furthermore, the retinal expression of mPPT occurs much earlier and at a higher level than in the brain at all developmental stages investigated. Since many retinal and brain proteins are highly palmitoylated and depalmitoylation by PPT is essential for their effective recycling in the lysosomes, our results raise the possibility that inactivating mutations of the PPT gene, as occur in INCL, are likely to cause cellular accumulation of lipid-modified proteins in the retina earlier than in the brain. Consequently, the loss of vision occurs before the deterioration of brain functions in this disease.


Asunto(s)
Ceguera/genética , Encéfalo/metabolismo , Lipofuscinosis Ceroideas Neuronales/genética , Retina/metabolismo , Tioléster Hidrolasas/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Ceguera/enzimología , Encéfalo/embriología , Mapeo Cromosómico , ADN Complementario , Humanos , Ratones , Datos de Secuencia Molecular , Lipofuscinosis Ceroideas Neuronales/enzimología , Retina/embriología
20.
Proc Natl Acad Sci U S A ; 96(7): 3963-8, 1999 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-10097146

RESUMEN

Uteroglobin (UG) is a multifunctional, secreted protein that has receptor-mediated functions. The human UG (hUG) gene is mapped to chromosome 11q12.2-13.1, a region frequently rearranged or deleted in many cancers. Although high levels of hUG expression are characteristic of the mucosal epithelia of many organs, hUG expression is either drastically reduced or totally absent in adenocarcinomas and in viral-transformed epithelial cells derived from the same organs. In agreement with these findings, in an ongoing study to evaluate the effects of aging on UG-knockout mice, 16/16 animals developed malignant tumors, whereas the wild-type littermates (n = 25) remained apparently healthy even after 11/2 years. In the present investigation, we sought to determine the effects of induced-expression of hUG in human cancer cells by transfecting several cell lines derived from adenocarcinomas of various organs with an hUG-cDNA construct. We demonstrate that induced hUG expression reverses at least two of the most important characteristics of the transformed phenotype (i.e., anchorage-independent growth on soft agar and extracellular matrix invasion) of only those cancer cells that also express the hUG receptor. Similarly, treatment of the nontransfected, receptor-positive adenocarcinoma cells with purified recombinant hUG yielded identical results. Taken together, these data define receptor-mediated, autocrine and paracrine pathways through which hUG reverses the transformed phenotype of cancer cells and consequently, may have tumor suppressor-like effects.


Asunto(s)
Adenocarcinoma/genética , Adenocarcinoma/patología , Transformación Celular Neoplásica , Cromosomas Humanos Par 11 , Uteroglobina/genética , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Mapeo Cromosómico , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Noqueados , Invasividad Neoplásica , Neoplasias Experimentales/genética , Fenotipo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética , Transfección , Células Tumorales Cultivadas , Neoplasias Uterinas/genética , Neoplasias Uterinas/patología , Uteroglobina/deficiencia , Uteroglobina/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA