Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Transl Med ; 22(1): 503, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38802941

RESUMEN

BACKGROUND: Prion diseases are transmissible and fatal neurodegenerative diseases characterized by accumulation of misfolded prion protein isoform (PrPSc), astrocytosis, microgliosis, spongiosis, and neurodegeneration. Elevated levels of cell membrane associated PrPSc protein and inflammatory cytokines hint towards the activation of death receptor (DR) pathway/s in prion diseases. Activation of DRs regulate, either cell survival or apoptosis, autophagy and necroptosis based on the adaptors they interact. Very little is known about the DR pathways activation in prion disease. DR3 and DR5 that are expressed in normal mouse brain were never studied in prion disease, so also their ligands and any DR adaptors. This research gap is notable and investigated in the present study. METHODS: C57BL/6J mice were infected with Rocky Mountain Laboratory scrapie mouse prion strain. The progression of prion disease was examined by observing morphological and behavioural abnormalities. The levels of PrP isoforms and GFAP were measured as the marker of PrPSc accumulation and astrocytosis respectively using antibody-based techniques that detect proteins on blot and brain section. The levels of DRs, their glycosylation and ectodomain shedding, and associated factors warrant their examination at protein level, hence western blot analysis was employed in this study. RESULTS: Prion-infected mice developed motor deficits and neuropathology like PrPSc accumulation and astrocytosis similar to other prion diseases. Results from this research show higher expression of all DR ligands, TNFR1, Fas and p75NTR but decreased levels DR3 and DR5. The levels of DR adaptor proteins like TRADD and TRAF2 (primarily regulate pro-survival pathways) are reduced. FADD, which primarily regulate cell death, its level remains unchanged. RIPK1, which regulate pro-survival, apoptosis and necroptosis, its expression and proteolysis (inhibits necroptosis but activates apoptosis) are increased. CONCLUSIONS: The findings from the present study provide evidence towards the involvement of DR3, DR5, DR6, TL1A, TRAIL, TRADD, TRAF2, FADD and RIPK1 for the first time in prion diseases. The knowledge obtained from this research discuss the possible impacts of these 16 differentially expressed DR factors on our understanding towards the multifaceted neuropathology of prion diseases and towards future explorations into potential targeted therapeutic interventions for prion disease specific neuropathology.


Asunto(s)
Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Enfermedades por Prión , Animales , Enfermedades por Prión/metabolismo , Enfermedades por Prión/patología , Receptores de Muerte Celular/metabolismo , Transducción de Señal , Encéfalo/metabolismo , Encéfalo/patología , Ratones , Proteínas PrPSc/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo
2.
Sci Rep ; 7: 41420, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28145533

RESUMEN

Brain expressed X-linked (Bex) genes are newer group of pro-apoptotic genes. Role of any Bex gene in neuroblastoma and Bex4 and Bex6 in any cancer is completely unknown. Re-expression of all endogenous Bex genes by any nutraceutical is also unknown. Therefore, we investigated the induction of all endogenous Bex genes and associated mechanisms by curcumin using N2a, an aggressive neuroblastoma cell line. Curcumin induced all endogenous Bex genes prior to apoptosis in N2a cells in a dose- and time-dependent manner. Wortmannin (PI-3Kinases inhibitor), SP600125 (JNK inhibitor) and pifithrin-α (p53 inhibitor) abrogated curcumin-mediated induction of Bex genes. Inhibition of curcumin-mediated induction of Bex genes by pifithrin-α also inhibited N2a cells apoptosis suggesting, a direct role of Bex genes in N2a cells apoptosis and involvement of p53 in Bex genes induction. Curcumin treatment activated p53 through hyperphosphorylation at serine 15 before Bex genes induction indicating Bex genes are novel downstream targets of p53. Collectively, curcumin, a safe nutraceutical has the potential to induce all endogenous Bex genes to harness their anti-cancer properties in neuroblastoma cells. Re-expression of Bex genes by curcumin acts as tumor suppressors and may provide alternate strategy to treat neuroblastomas and other cancers with silenced Bex genes.


Asunto(s)
Apoptosis/genética , Curcumina/farmacología , Regulación Neoplásica de la Expresión Génica/genética , Proteínas del Tejido Nervioso/genética , Neuroblastoma/genética , Neuroblastoma/patología , Proteína p53 Supresora de Tumor/metabolismo , Androstadienos/farmacología , Animales , Antracenos/farmacología , Apoptosis/efectos de los fármacos , Benzotiazoles/farmacología , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Modelos Biológicos , Proteínas del Tejido Nervioso/metabolismo , Neuroblastoma/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Tolueno/análogos & derivados , Tolueno/farmacología , Wortmanina
3.
Springerplus ; 3: 161, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25140287

RESUMEN

Increased production, oligomerization and aggregation of amyloid-ß (Aß) peptides are hallmark pathologies of Alzheimer's disease (AD). Expressing familial AD mutations (amyloid precursor protein and/or presenilins mutations), the Aß-pathologies of AD has been recapitulated in animal models of AD. Very few primary cell culture-based models of AD are available and they exhibit very weak Aß-pathologies compared to what is seen in AD patients and animal models of AD. CNS stem/progenitor cells are present in both embryonic and adult brains. They can be isolated, grown as neurospheres and differentiated into neurons, astrocytes and oligodendrocytes. It is not yet known whether CNS stem/progenitor cells can support the production of Aß peptides in culture. In this report, we have established Aß-pathologies such as production, secretion, oligomerization and aggregation of Aß peptides utilizing neurosphere cultures to create a new cellular model of AD. These cultures were developed from E15 embryonic brains of transgenic mice carrying the Swedish mutations in humanized mouse APP cDNA and the exon-9 deleted human presenilin 1 cDNA both regulated by mouse prion protein gene (Prnp) promoter. Results demonstrated the expression of transgene transcripts, APPswe protein and its processed products only in transgene positive neurosphere cultures. These cultures generate and secrete both Aß40 and Aß42 peptides into culture medium at levels comparable to the Aß load in the brain of AD patients and animal models of AD, and produce pathogenic oligomers of Aß peptides. The Aß42/Aß40 ratio in the medium of transgene positive neurosphere cultures is higher than any known cellular models of AD. Conformation dependent immunocytochemistry demonstrated the possible presence of intracellular and extracellular aggregation of Aß peptides in neurosphere cultures, which are also seen in AD brain and animal models of AD. Collectively, our neurosphere cultures provide robust Aß-pathologies of AD better than existing cellular model of Alzheimer's disease.

4.
PLoS One ; 8(6): e66430, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23824039

RESUMEN

Neuroblastoma is the most common cancer in infants and fourth most common cancer in children. Despite recent advances in cancer treatments, the prognosis of stage-IV neuroblastoma patients continues to be dismal which warrant new pharmacotherapy. A novel tetracyclic condensed quinoline compound, 8-methoxypyrimido [4',5':4,5]thieno(2,3-b) quinoline-4(3H)-one (MPTQ) is a structural analogue of an anticancer drug ellipticine and has been reported to posses anticancer property. Study on MPTQ on neuroblastoma cells is very limited and mechanisms related to its cytotoxicity on neuroblastoma cells are completely unknown. Here, we evaluated the anticancer property of MPTQ on mouse neuro 2a and human SH-SY5Y neuroblastoma cells and investigated the mechanisms underlying MPTQ-mediated neuro 2a cell death. MPTQ-mediated neuro 2a and SH-SY5Y cell deaths were found to be dose and time dependent. Moreover, MPTQ induced cell death reached approximately 99.8% and 90% in neuro 2a and SH-SY5Y cells respectively. Nuclear oligonucleosomal DNA fragmentation and Terminal dUTP Nick End Labelling assays indicated MPTQ-mediated neuro 2a cell death involved apoptosis. MPTQ-mediated apoptosis is associated with increased phosphorylation of p53 at Ser15 and Ser20 which correlates with the hyperphosphorylation of Ataxia-Telangiectasia mutated protein (ATM). Immunocytochemical analysis demonstrated the increased level of Bax protein in MPTQ treated neuro 2a cells. MPTQ-mediated apoptosis is also associated with increased activation of caspase-9, -3 and -7 but not caspase-2 and -8. Furthermore, increased level of caspase-3 and cleaved Poly (ADP Ribose) polymerase were observed in the nucleus of MPTQ treated neuro 2a cells, suggesting the involvement of caspase-dependent intrinsic but not extrinsic apoptotic pathway. Increased nuclear translocation of apoptosis inducing factor suggests additional involvement of caspase-independent apoptosis pathway in MPTQ treated neuro 2a cells. Collectively, MPTQ-induced neuro 2a cell death is mediated by ATM and p53 activation, and Bax-mediated activation of caspase-dependent and caspase-independent mitochondrial apoptosis pathways.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Neuroblastoma/patología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Neuroblastoma/enzimología , Neuroblastoma/metabolismo
5.
Mol Carcinog ; 52(6): 413-25, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22213363

RESUMEN

Polycyclic aromatic molecules such as ellipticine intercalate into double-stranded DNA and interfere with physiological functions. In the present study, we evaluate the chemotherapeutic potential of MPTQ on animal models and its mode of action. In order to test the antitumor activity, monohydrochloride of MPTQ was orally administered in mice bearing tumor. Results showed a significant inhibition of tumor growth compared to that of untreated controls. More importantly, mean lifespan of tumor bearing animals treated with MPTQ was significantly higher as compared to that of untreated tumor bearing mice suggesting that the treatment affected viability of cancerous cells, but not of normal cells. Consistent with this, we find that administration of MPTQ to normal mice did not cause any major side effects as observed upon hematological and serum profiling. We also found that MPTQ induces cytotoxicity in cancer cell lines, by activating apoptosis both by intrinsic and extrinsic pathways. Thus, MPTQ could be used as a potential cancer therapeutic agent.


Asunto(s)
Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Sustancias Intercalantes/uso terapéutico , Neoplasias/tratamiento farmacológico , Quinolinas/uso terapéutico , Tiofenos/uso terapéutico , Animales , Antineoplásicos/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , ADN/genética , Roturas del ADN de Doble Cadena/efectos de los fármacos , Humanos , Sustancias Intercalantes/farmacología , Leucemia/tratamiento farmacológico , Leucemia/genética , Leucemia/metabolismo , Leucemia/patología , Ratones , Ratones Endogámicos BALB C , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Quinolinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Tiofenos/farmacología
6.
Proc Natl Acad Sci U S A ; 103(10): 3875-80, 2006 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-16495413

RESUMEN

Only a few cell lines have been infected with prions, offering limited genetic diversity and sensitivity to several strains. Here we report that cultured neurospheres expressing cellular prion protein (PrP(C)) can be infected with prions. Neurosphere lines isolated from the brains of mice at embryonic day 13-15 grow as aggregates and contain CNS stem cells. We produced neurosphere cultures from FVB/NCr (FVB) mice, from transgenic (Tg) FVB mice that overexpress mouse PrP-A (Tg4053), and from congenic FVB mice with a targeted null mutation in the PrP gene (Prnp(0/0)) and incubated them with the Rocky Mountain Laboratory prion strain. While monitoring the levels of disease-causing PrP (PrP(Sc)) at each passage, we observed a dramatic rise in PrP(Sc) levels with time in the Tg4053 neurosphere cells, whereas the level of PrP(Sc) decayed to undetectable levels in cell cultures lacking PrP. PrP(Sc) levels in cultures from FVB mice initially declined but then increased with passage. Prions produced in culture were transmissible to mice and produced disease pathology. Intracellular aggregates of PrP(Sc) were present in cells from infected cultures. The susceptibility of neurosphere cultures to prions mirrored that of the mice from which they were derived. Neurosphere lines from Tg4053 mice provide a sensitive in vitro bioassay for mouse prions; neurosphere lines from other Tg mice overexpressing PrP might be used to assay prions from other species, including humans.


Asunto(s)
Neuronas/metabolismo , Priones/patogenicidad , Animales , Bioensayo , Expresión Génica , Ratones , Ratones Noqueados , Ratones Transgénicos , Complejos Multiproteicos , Mutación , Proteínas PrPC/genética , Proteínas PrPC/metabolismo , Proteínas PrPC/patogenicidad , Proteínas PrPSc/química , Proteínas PrPSc/genética , Proteínas PrPSc/metabolismo , Proteínas PrPSc/patogenicidad , Enfermedades por Prión/genética , Enfermedades por Prión/metabolismo , Priones/genética , Priones/metabolismo , Esferoides Celulares
7.
Am J Physiol Cell Physiol ; 289(2): C264-76, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15743889

RESUMEN

In Alzheimer's disease (AD), one finds increased presence of monocytes/macrophages and activated microglial cells in the brain. Immunohistochemical studies show increased expression of chemokine receptor 5 (CCR5) on reactive microglia associated with amyloid deposits in AD, suggesting that CCR5 may play a role in the regulation of the immune response in AD. In this study, we used peripheral blood monocytes and human monocytic THP-1 cell line as a model of microglia to delineate the cellular signaling mechanism of Abeta-induced CCR5 expression and the latter's role in the chemotaxis of monocytes. We observed that Abeta peptides at pathophysiological concentrations (125 nM) increased CCR5 mRNA and cell surface protein expression. The cellular signaling involved activation of c-Raf, ERK-1/ERK-2, and c-Jun NH(2)-terminal kinase. Analysis of some transcription factors associated with CCR5 promoter revealed that Abeta increased DNA binding activity of Egr-1 and AP-1. In addition, we show that CCR5 promoter contains an Egr-1 like consensus sequence GCGGGGGTG as demonstrated by 1) electrophoretic mobility shift assay, 2) transfection studies with truncated CCR5 gene promoter construct, and 3) chromatin immunoprecipitation analysis. Moreover, transfection of Egr-1 siRNA, but not of scrambled Egr-1 siRNA, in THP-1 cells resulted in >75% reduction in both Abeta-mediated CCR5 expression and concomitant chemotaxis to its ligands. We suggest that inhibition of Egr-1 by either Egr-1 siRNA or pharmacological agents may reduce activation of monocytes/microglia and possibly ameliorate the inflammation and progression of AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , ADN de Cadena Simple/genética , Monocitos/metabolismo , ARN Interferente Pequeño/metabolismo , Receptores CCR5/biosíntesis , Transducción de Señal/fisiología , Secuencia de Bases , Línea Celular , Quimiotaxis de Leucocito/fisiología , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Citometría de Flujo , Expresión Génica , Regulación de la Expresión Génica , Humanos , Modelos Biológicos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Receptores CCR5/genética , Transfección , Quinasas raf/metabolismo
8.
J Neurochem ; 91(5): 1199-210, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15569263

RESUMEN

Epidemiological studies show reduced risk of Alzheimer's disease (AD) among patients using non-steroidal inflammatory drugs (NSAID) indicating the role of inflammation in AD. Studies have shown a chronic CNS inflammatory response associated with increased accumulation of amyloid peptide and activated microglia in AD. Our previous studies showed that interaction of Abeta1-40 or fibrilar Abeta1-42 caused activation of nuclear transcription factor, early growth response-1 (Egr-1), which resulted in increased expression of cytokines (TNF-alpha and IL-1beta) and chemokines (MIP-1beta, MCP-1 and IL-8) in monocytes. We determined whether curcumin, a natural product known to have anti-inflammatory properties, suppressed Egr-1 activation and concomitant expression of cytochemokines. We show that curcumin (12.5-25 microm) suppresses the activation of Egr-1 DNA-binding activity in THP-1 monocytic cells. Curcumin abrogated Abeta1-40-induced expression of cytokines (TNF-alpha and IL-1beta) and chemokines (MIP-1beta, MCP-1 and IL-8) in both peripheral blood monocytes and THP-1 cells. We found that curcumin inhibited Abeta1-40-induced MAP kinase activation and the phosphorylation of ERK-1/2 and its downstream target Elk-1. We observed that curcumin inhibited Abeta1-40-induced expression of CCR5 but not of CCR2b in THP-1 cells. This involved abrogation of Egr-1 DNA binding in the promoter of CCR5 by curcumin as determined by: (i) electrophoretic mobility shift assay, (ii) transfection studies with truncated CCR5 gene promoter constructs, and (iii) chromatin immunoprecipitation analysis. Finally, curcumin inhibited chemotaxis of THP-1 monocytes in response to chemoattractant. The inhibition of Egr-1 by curcumin may represent a potential therapeutic approach to ameliorate the inflammation and progression of AD.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Quimiotaxis/efectos de los fármacos , Curcuma/química , Curcumina/farmacología , Citocinas/metabolismo , Expresión Génica/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Receptores CCR5/metabolismo , Western Blotting/métodos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cromatina/metabolismo , Citocinas/genética , Citocinas/farmacología , Proteínas de Unión al ADN/metabolismo , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Proteína 1 de la Respuesta de Crecimiento Precoz , Ensayo de Cambio de Movilidad Electroforética/métodos , Inhibidores Enzimáticos/farmacología , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Inmunoprecipitación/métodos , Monocitos/efectos de los fármacos , Monocitos/fisiología , Oligodesoxirribonucleótidos Antisentido/farmacología , ARN Mensajero/metabolismo , Receptores CCR5/genética , Factores de Transcripción/metabolismo , Transfección/métodos
9.
Liver Int ; 24(4): 361-70, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15287860

RESUMEN

BACKGROUND: The availability of well-characterized human liver cell populations that can be frozen and thawed will be critical for cell therapy. We addressed whether human hepatocytes can recover after cryopreservation and engraft in immunodeficient mice. METHODS: We isolated cells from discarded human livers and studied the properties of cryopreserved cells. The viability of thawed cells was established with multiple in vitro assays, including analysis of liver gene expression, ureagenesis, cytochrome P450 activity, and growth factor-induced cell proliferation. The fate of transplanted cells was analysed in immunodeficient NOD-SCID mice. RESULTS: After thawing, the viability of human hepatocytes exceeded 60%. Cells attached to culture dishes, proliferated following growth factor stimulation and exhibited liver-specific functions. After transplantation in NOD-SCID mice, cells engrafted in the peritoneal cavity, a heterologous site, as well as the liver itself, retained hepatic function and proliferated in response to liver injury. Transplanted hepatocytes were integrated in the liver parenchyma. Occasionally, transplanted cells were integrated in bile ducts. CONCLUSIONS: Cryopreserved human liver cell showed the ability to retain functional integrity and to reconstitute both hepatic and biliary lineages in mice. These studies offer suitable paradigms aimed at characterizing liver cells prior to transplantation in people.


Asunto(s)
Criopreservación , Supervivencia de Injerto , Hepatocitos/citología , Hepatocitos/trasplante , Animales , Bioensayo/métodos , División Celular , Movimiento Celular , Supervivencia Celular , Humanos , Hígado/citología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante Heterólogo
10.
Hum Mol Genet ; 13(18): 1989-97, 2004 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-15254013

RESUMEN

Phenotypes produced by expression of human amyloid precursor protein (APP) transgenes vary depending on the genetic background of the mouse. FVB/N mice overexpressing human APP695 develop a central nervous system disorder and die prematurely, precluding development of Abeta peptide amyloid plaques. 129S6 mice are resistant to the lethal effects of APP overexpression, allowing sufficient levels of Abeta expression for the development of amyloid plaques and age-dependent memory deficits. To identify the genes that determine susceptibility or resistance to APP we analyzed crosses involving FVB/NCr and 129S6.Tg2576 mice that overexpress 'Swedish' mutant (K670N, M671L) APP695. APP transgene-positive FVB129S6F1 (F1) mice are resistant to the lethal effects of APP overexpression, so FVBxF1 backcross and F2 intercross offspring were produced. Analysis of age of death as a quantitative trait revealed significant linkage to loci on proximal chromosome 14 and on chromosome 9; 129S6 alleles protect against the lethal effects of APP. Within the chromosome 14 interval are segments homologous to regions on human chromosome 10 that have been linked to late onset Alzheimer's disease or to levels of Abeta peptide in plasma. However, analysis of plasma Abeta peptide concentrations at 6 weeks in backcross offspring produced no significant linkage. Similarly, elevation of human Abeta peptide concentrations by expression of mutant presenilin transgenes did not increase the proportion of mice dying prematurely, suggesting that early death reflects effects of APP or fragments other than Abeta.


Asunto(s)
Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Genes Letales/genética , Predisposición Genética a la Enfermedad , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/sangre , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Mapeo Cromosómico , Humanos , Ratones , Ratones Mutantes , Fragmentos de Péptidos/sangre , Fenotipo , Transgenes/genética
11.
Exp Cell Res ; 291(2): 451-62, 2003 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-14644166

RESUMEN

To investigate whether oxidative manipulation of extracellular matrix components could affect cell survival, we studied primary rat hepatocytes cultured on dishes coated with collagen type 1, which was oxidized with a metal-based system. Culture of hepatocytes on oxidized collagen led to decreased cellular catalase activity along with impaired cell survival. The fraction of polyploid hepatocytes decreased early followed by greater reaccumulation of polyploid cells. Cells cultured on oxidized collagen showed greater susceptibility to additional oxidant stress induced by tert.-butyl-hydroperoxide. The capacity of hepatocytes for growth factor-induced DNA synthesis was unaffected by culture on oxidized collagen. In response to culture on oxidized matrix, AP-1, Egr-1, CREB, and NF-kappaB transcription factor activity was rapidly increased. This change in transcription factor activity was ameliorated by treatment of collagen with a free radical spin trap, N-tert.-butyl-alpha-phenylnitrone, prior to oxidation. Moreover, culture of hepatocytes with aminoguanidine, an antioxidant drug, decreased cell injury. These findings established that exposure of primary hepatocytes to oxidized extracellular matrix components rapidly activates cell signaling events with loss of hepatocyte subpopulations. Such cell-extracellular matrix interactions may play roles in organ homeostasis and oncogenetic progression.


Asunto(s)
Colágeno Tipo I/metabolismo , Colágeno Tipo I/fisiología , Hepatocitos/citología , Transducción de Señal , Animales , Catálisis , Supervivencia Celular , Células Cultivadas , Matriz Extracelular/metabolismo , Matriz Extracelular/fisiología , Metales , Oxidación-Reducción , Estrés Oxidativo , Ratas , Ratas Endogámicas F344 , Factores de Transcripción/metabolismo , terc-Butilhidroperóxido
12.
J Immunol ; 170(10): 5281-94, 2003 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-12734378

RESUMEN

In Alzheimer's disease (AD) one finds increased deposition of A beta and also an increased presence of monocytes/macrophages in the vessel wall and activated microglial cells in the brain. AD patients show increased levels of proinflammatory cytokines by activated microglia. Here we used a human monocytic THP-1 cell line as a model for microglia to delineate the cellular signaling mechanism involved in amyloid peptides (A beta(1-40) and A beta(1-42))-induced expression of inflammatory cytokines and chemokines. We observed that A beta peptides at physiological concentrations (125 nM) increased mRNA expression of cytokines (TNF-alpha, and IL-1 beta) and chemokines (monocyte chemoattractant protein-1 (MCP-1), IL-8, and macrophage inflammatory protein-1 beta (MIP-1 beta)). The cellular signaling involved activation of c-Raf, extracellular signal-regulated kinase-1 (ERK-1)/ERK-2, and c-Jun N-terminal kinase, but not p38 mitogen-activated protein kinase. This is further supported by the data showing that A beta causes phosphorylation of ERK-1/ERK-2, which, in turn, activates Elk-1. Furthermore, A beta mediated a time-dependent increase in DNA binding activity of early growth response-1 (Egr-1) and AP-1, but not of NF-kappa B and CREB. Moreover, A beta-induced Egr-1 DNA binding activity was reduced >60% in THP-1 cells transfected with small interfering RNA duplexes for Egr-1 mRNA. We show that A beta-induced expression of TNF-alpha, IL-1 beta, MCP-1, IL-8, and MIP-1 beta was abrogated in Egr-1 small inhibitory RNA-transfected cells. Our results indicate that A beta-induced expression of cytokines (TNF-alpha and IL-1 beta) and chemokines (MCP-1, IL-8, and MIP-1 beta) in THP-1 monocytes involves activation of ERK-1/ERK-2 and downstream activation of Egr-1. The inhibition of Egr-1 by Egr-1 small inhibitory RNA may represent a potential therapeutic target to ameliorate the inflammation and progression of AD.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Quimiocinas/biosíntesis , Citocinas/biosíntesis , Proteínas de Unión al ADN/fisiología , Monocitos/metabolismo , Ácidos Nucleicos Heterodúplex/fisiología , Fragmentos de Péptidos/farmacología , Proteínas Serina-Treonina Quinasas , ARN Mensajero/fisiología , ARN Interferente Pequeño/fisiología , Factores de Transcripción/fisiología , Péptidos beta-Amiloides/antagonistas & inhibidores , Antracenos/farmacología , Células Cultivadas , Quimiocinas/antagonistas & inhibidores , Quimiocinas/sangre , Quimiocinas/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Citocinas/antagonistas & inhibidores , Citocinas/sangre , Citocinas/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz , Flavonoides/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/fisiología , Monocitos/enzimología , Monocitos/inmunología , FN-kappa B/metabolismo , Fragmentos de Péptidos/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-raf/fisiología , ARN Mensajero/biosíntesis , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional/efectos de los fármacos , Células Tumorales Cultivadas
13.
Proc Natl Acad Sci U S A ; 100(12): 7253-8, 2003 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-12756298

RESUMEN

Beta-cell replacement is considered to be the most promising approach for treatment of type 1 diabetes. Its application on a large scale is hindered by a shortage of cells for transplantation. Activation of insulin expression, storage, and regulated secretion in stem/progenitor cells offers novel ways to overcome this shortage. We explored whether fetal human progenitor liver cells (FH) could be induced to differentiate into insulin-producing cells after expression of the pancreatic duodenal homeobox 1 (Pdx1) gene, which is a key regulator of pancreatic development and insulin expression in beta cells. FH cells possess a considerable replication capacity, and this was further extended by introduction of the gene for the catalytic subunit of human telomerase. Immortalized FH cells expressing Pdx1 activated multiple beta-cell genes, produced and stored considerable amounts of insulin, and released insulin in a regulated manner in response to glucose. When transplanted into hyperglycemic immunodeficient mice, the cells restored and maintained euglycemia for prolonged periods. Quantitation of human C-peptide in the mouse serum confirmed that the glycemia was normalized by the transplanted human cells. This approach offers the potential of a novel source of cells for transplantation into patients with type 1 diabetes.


Asunto(s)
Hepatocitos/trasplante , Proteínas de Homeodominio , Hiperglucemia/terapia , Insulina/biosíntesis , Trasplante de Células Madre/métodos , Animales , Glucemia/metabolismo , Péptido C/sangre , Diferenciación Celular , Proteínas de Unión al ADN , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/terapia , Femenino , Expresión Génica , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Hiperglucemia/sangre , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre/citología , Células Madre/metabolismo , Telomerasa/genética , Transactivadores/genética , Transducción Genética , Trasplante Heterólogo
14.
Blood ; 102(4): 1515-24, 2003 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-12689930

RESUMEN

Monocytes from patients with sickle cell disease (SCD) are in an activated state. However, the mechanism of activation of monocytes in SCD is not known. Our studies showed that placenta growth factor (PlGF) activated monocytes and increased mRNA levels of cytokines (tumor necrosis factor-alpha [TNF-alpha] and interleukin-1beta [IL-1beta]) and chemokines (monocyte chemotactic protein-1 [MCP-1], IL-8, and macrophage inflammatory protein-1beta [MIP-1beta]) in both normal monocytes and in the THP-1 monocytic cell line. This increase in mRNA expression of cytochemokines was also reflected in monocytes derived from subjects with SCD. We studied the PlGF-mediated downstream cellular signaling events that caused increased transcription of inflammatory cytochemokines and chemotaxis of THP-1 monocytes. PlGF-mediated cytochemokine mRNA and protein expression was inhibited by PD98059 and wortmannin, inhibitors of mitogen-activated protein kinase kinase (MAPK/MEK) kinase and phosphatidylinositol-3 (PI3) kinase, respectively, but not by SB203580, a p38 kinase inhibitor. PlGF caused a time-dependent transient increase in phosphorylation of extracellular signal-regulated kinase-1/2 (ERK-1/2), which was completely inhibited by wortmannin, indicating that activation of PI3 kinase preceded MEK activation. PlGF also induced transient phosphorylation of AKT. MEK and PI3 kinase inhibitors and antibody to Flt-1 abrogated PlGF-induced chemotaxis of THP-1 monocytes. Overexpression of a dominant-negative AKT or a dominant-negative PI3 kinase p85 subunit in THP-1 monocytes attenuated the PlGF-mediated phosphorylation of ERK-1/2, cytochemokine secretion, and chemotaxis. Taken together, these data show that activation of monocytes by PlGF occurs via activation of Flt-1, which results in activation of PI3 kinase/AKT and ERK-1/2 pathways. Therefore, we propose that increased levels of PlGF in circulation play an important role in the inflammation observed in SCD via its effects on monocytes.


Asunto(s)
Quimiocinas/biosíntesis , Citocinas/biosíntesis , Monocitos/metabolismo , Proteínas Gestacionales/farmacología , Anemia de Células Falciformes/sangre , Células Cultivadas , Quimiocinas/sangre , Quimiocinas/genética , Quimiocinas/metabolismo , Quimiotaxis/fisiología , Citocinas/sangre , Citocinas/genética , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Expresión Génica/efectos de los fármacos , Humanos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Monocitos/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Factor de Crecimiento Placentario , Proteínas Gestacionales/antagonistas & inhibidores , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factores de Tiempo , Transfección , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
15.
Gastroenterology ; 124(2): 432-44, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12557149

RESUMEN

BACKGROUND & AIMS: The availability of in vitro expandable human hepatocytes would greatly advance liver-directed cell therapies. Therefore, we examined whether human fetal hepatocytes are amenable to telomerase-mediated immortalization without inducing a transformed phenotype and disrupting their differentiation potential. Telomerase is a ribonucleoprotein that plays a pivotal role in maintaining telomere length and chromosome stability. Human somatic cells, including hepatocytes, exhibit no telomerase activity. Consequently, their telomeres progressively shorten with each cell cycle until critically short telomeres trigger replicative senescence. METHODS: The catalytic subunit, telomerase reverse transcriptase, was expressed in human fetal hepatocytes. Transduced cells were characterized for telomerase activity, telomere length, proliferative capacity, hepatocellular functions, oncogenicity, and their in vivo maturation potential. RESULTS: The expression of human telomerase reverse transcriptase restored telomerase activity in human fetal hepatocytes. Telomerase-reconstituted cells were capable of preserving elongated telomeres, propagated in culture beyond replicative senescence for more than 300 cell doublings (to date), and maintained their liver-specific nature, as analyzed by a panel of hepatic growth factors, growth factor receptors, and transcription factors as well as albumin, glucose-6-phosphatase, glycogen synthesis, cytochrome P450 (CYP) expression profiles, and urea production. Moreover, the immortalized cells exhibited no oncogenicity, and no up-regulation of c-Myc was detected. The cells engrafted and survived in the liver of immunodeficient mice with hepatocellular gene expression. CONCLUSIONS: Reconstitution of telomerase activity induces indefinite replication in human fetal hepatocytes and offers unique opportunities for examining basic biologic mechanisms and for considering development of stable cell lines for liver-directed therapies.


Asunto(s)
Feto/citología , Hepatocitos/fisiología , Telomerasa/metabolismo , Animales , Diferenciación Celular , Línea Celular Transformada/fisiología , Proteínas de Unión al ADN , Femenino , Hepatocitos/citología , Hepatocitos/enzimología , Humanos , Ratones , Neoplasias/etiología , Embarazo , Telomerasa/genética , Telómero/ultraestructura , Transducción Genética
16.
Invest Ophthalmol Vis Sci ; 44(1): 22-31, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12506051

RESUMEN

PURPOSE: To devise methods for unequivocal identification of activated retinal microglia in experimental autoimmune uveoretinitis (EAU) and to investigate their role in the development of EAU. METHODS: A group of Lewis rats underwent optic nerve axotomy with the application of N-4-(4-didecylaminostyryl)-N methylpyridinium iodide (4Di-10ASP) at the axotomy site. On days 3, 14, and 38 after axotomy, the rats were killed, the eyes were enucleated, and the retinas were stained for OX42. Another group of such axotomized rats were immunized with S-antigen peptide and were killed on days 7 through 12 after the injection with peptide. The enucleated eyes were stained for OX42 and examined by confocal microscope. After axotomy, bone marrow (Y-->X) chimeric rats were injected with S-antigen peptide and were killed on days 10 and 12 after injection. The retinas were evaluated by PCR with Y-specific primers. Finally, a group of axotomized rats was injected with the S-antigen peptide and killed on days 6, 8, 9, and 10 after injection. Their enucleated eyes were examined for microglial expression of TNFalpha and for generation of peroxynitrite. RESULTS: In the axotomized, non-EAU eyes, 4Di-10ASP-labeled ganglion cells were detectable on days 3 and 14, and 4Di-10ASP-containing OX42-positive cells (microglia) were found in the nerve fiber and other inner retinal layers on days 14 and 38. The S-antigen peptide-injected rats showed migration of the microglia (4Di-10ASP-positive and OX42-positive) to the photoreceptor cell layer on day 9, and these cells increased in number at this site on day 10. No macrophages (OX42-positive and 4Di-10ASP-negative) were present at this early stage of EAU, but such cells appeared in the retina on days 11 and 12. PCR of the chimeric EAU retinas showed an absence of the Y chromosome-amplified product on day 10, but the presence of this product was detected on day 12. The expression of TNFalpha and generation of peroxynitrite were noted in the migrated microglia at the photoreceptor cell layer on days 9 and 10 of EAU. CONCLUSIONS: In the early phase of EAU, the microglia migrate to the photoreceptor cell layer where they generate TNFalpha and peroxynitrite. Such microglial migration and activation take place before infiltration of the macrophages. These findings indicate a novel pathogenic mechanism of EAU, in which retinal microglia may initiate retinitis with subsequent recruitment of circulation-derived phagocytes, leading to the amplification of uveoretinitis.


Asunto(s)
Microglía/fisiología , Células Ganglionares de la Retina/fisiología , Retinitis/patología , Uveítis/patología , Animales , Arrestina , Axotomía , Movimiento Celular/fisiología , Quimera , Modelos Animales de Enfermedad , Femenino , Colorantes Fluorescentes , Masculino , Microscopía Confocal , Nervio Óptico/fisiología , Ácido Peroxinitroso/metabolismo , Reacción en Cadena de la Polimerasa , Compuestos de Piridinio , Ratas , Ratas Endogámicas Lew , Retinitis/inducido químicamente , Retinitis/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Uveítis/inducido químicamente , Uveítis/metabolismo , Cromosoma Y/metabolismo
17.
Am J Physiol Cell Physiol ; 283(3): C895-904, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12176746

RESUMEN

During normal aging and amyloid beta-peptide (Abeta) disorders such as Alzheimer's disease (AD), one finds increased deposition of Abeta and activated monocytes/microglial cells in the brain. Our previous studies show that Abeta interaction with a monolayer of normal human brain microvascular endothelial cells results in increased adherence and transmigration of monocytes. Relatively little is known of the role of Abeta accumulated in the AD brain in mediating trafficking of peripheral blood monocytes (PBM) across the blood-brain barrier (BBB) and concomitant accumulation of monocytes/microglia in the AD brain. In this study, we showed that interaction of Abeta(1--40) with apical surface of monolayer of brain endothelial cells (BEC), derived either from normal or AD individuals, resulted in increased transendothelial migration of monocytic cells (HL-60 and THP-1) and PBM. However, transmigration of monocytes across the BEC monolayer cultivated in a Transwell chamber was increased 2.5-fold when Abeta was added to the basolateral side of AD compared with normal individual BEC. The Abeta-induced transmigration of monocytes was inhibited in both normal and AD-BEC by antibodies to the putative Abeta receptor, receptor for advanced glycation end products (RAGE), and to the endothelial cell junction molecule, platelet-endothelial cell adhesion molecule-1 (PECAM-1). We conclude that interaction of Abeta with the basolateral surface of AD-BEC induces cellular signaling, promoting transmigration of monocytes from the apical to basolateral direction. We suggest that Abeta in the AD brain parenchyma or cerebrovasculature initiates cellular signaling that induces PBM to transmigrate across the BBB and accumulate in the brain.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/farmacología , Polaridad Celular , Endotelio Vascular/metabolismo , Monocitos/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Anciano , Anciano de 80 o más Años , Anticuerpos/farmacología , Encéfalo/irrigación sanguínea , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Cámaras de Difusión de Cultivos , Endotelio Vascular/citología , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Masculino , Monocitos/citología , Fosforilación/efectos de los fármacos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/antagonistas & inhibidores , Receptores Inmunológicos/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...