Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Neuron ; 54(3): 379-86, 2007 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-17481392

RESUMO

Thermosensation is an essential sensory function that is subserved by a variety of transducer molecules, including those from the Transient Receptor Potential (TRP) ion channel superfamily. One of its members, TRPM8 (CMR1), a ligand-gated, nonselective cation channel, is activated by both cold and chemical stimuli in vitro. However, its roles in cold thermosensation and pain in vivo have not been fully elucidated. Here, we show that sensory neurons derived from TRPM8 null mice lack detectable levels of TRPM8 mRNA and protein and that the number of these neurons responding to cold (18 degrees C) and menthol (100 microM) is greatly decreased. Furthermore, compared with WT mice, TRPM8 null mice display deficiencies in certain behaviors, including icilin-induced jumping and cold sensation, as well as a significant reduction in injury-induced responsiveness to acetone cooling. These results suggest that TRPM8 may play an important role in certain types of cold-induced pain in humans.


Assuntos
Temperatura Baixa , Camundongos Knockout/fisiologia , Canais de Cátion TRPM/deficiência , Sensação Térmica/genética , Análise de Variância , Animais , Comportamento Animal/efeitos dos fármacos , Cálcio/metabolismo , Capsaicina/farmacologia , Células Cultivadas , Gânglios Espinais/citologia , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/fisiopatologia , Metanol/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos dos fármacos , Atividade Motora/genética , Neurônios Aferentes/efeitos dos fármacos , Neurônios Aferentes/fisiologia , Medição da Dor/métodos , Pirimidinonas/farmacologia , Tempo de Reação/efeitos dos fármacos
2.
BMC Genomics ; 12: 464, 2011 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-21943378

RESUMO

BACKGROUND: The creation of lymphoblastoid cell lines (LCLs) through Epstein-Barr virus (EBV) transformation of B-lymphocytes can result in a valuable biomaterial for cell biology research and a renewable source of DNA. While LCLs have been used extensively in cellular and genetic studies, the process of cell transformation and expansion during culturing may introduce genomic changes that may impact their use and the interpretation of subsequent genetic findings. RESULTS: We performed whole exome sequencing on a tetrad family using DNA derived from peripheral blood mononuclear cells (PBMCs) and LCLs from each individual. We generated over 4.7 GB of mappable sequence to a 125X read coverage per sample. An average of 19,354 genetic variants were identified. Comparison of the two DNA sources from each individual showed an average concordance rate of 95.69%. By lowering the variant calling parameters, the concordance rate between the paired samples increased to 99.82%. Sanger sequencing of a subset of the remaining discordant variants did confirm the presence of de novo mutations arising in LCLs. CONCLUSIONS: By varying software stringency parameters, we identified 99% concordance between DNA sequences derived from the two different sources from the same donors. These results suggest that LCLs are an appropriate representation of the genetic material of the donor and suggest that EBV transformation can result in low-level generation of de novo mutations. Therefore, use of PBMC or early passage EBV-transformed cells is recommended. These findings have broad-reaching implications, as there are thousands of LCLs in public biorepositories and individual laboratories.


Assuntos
Linfócitos B/virologia , Transformação Celular Viral , DNA/química , Herpesvirus Humano 4/fisiologia , Leucócitos Mononucleares/metabolismo , Linfócitos B/fisiologia , Doadores de Sangue , Linhagem Celular Transformada , Transformação Celular Viral/genética , DNA/metabolismo , Exoma/genética , Humanos , Análise de Sequência de DNA
3.
Int J Nanomedicine ; 14: 6451-6464, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31496697

RESUMO

BACKGROUND: We recently reported on long-term comprehensive biocompatibility and biodistribution study of fluorescent nanodiamond particles (NV)-Z-average 800nm (FNDP-(NV)) in rats. FNDP-(NV) primary deposition was found in the liver, yet liver function tests remained normal. PURPOSE: The present study aimed to gain preliminary insights on discrete localization of FNDP-(NV) in liver cells of the hepatic lobule unit and venous micro-vasculature. Kinetics of FDNP-(NV) uptake into liver cells surrogates in culture was conducted along with cell cytokinesis as markers of cells' viability. METHODS: Preserved liver specimens from a pilot consisting of two animals which were stained for cytoskeletal elements (fluorescein-isothiocyanate-phalloidin) were examined for distribution of FNDP-(NV) by fluorescent microscopy (FM) and Confocal-FM (CFM) using near infra-red fluorescence (NIR). Hepatocellular carcinoma cells (HepG-2) and human umbilical vein endothelial cells (HUVEC) were cultured with FNDP-(NV) and assayed for particle uptake and location using spectrophotometric technology and microscopy. RESULTS: HepG-2 and HUVEC displayed rapid (<30 mins) onset and concentration-dependent FNDP-(NV) internalization and formation of peri-nuclear corona. FM/CFM of liver sections revealed FNDP-(NV) presence throughout the hepatic lobules structures marked by spatial distribution, venous microvascular spaces and parenchyma and non-parenchyma cells. CONCLUSION: The robust presence of FNDP-(NV) throughout the hepatic lobules including those internalized within parenchyma cells and agglomerates in the liver venous micro-circulation were not associated with macro or micro histopathological signs nor vascular lesions. Cells cultures indicated normal cytokinesis in cells containing FNDP-(NV) agglomerates. Liver parenchyma cells and the liver microcirculation remain agnostic to presence of FNDP-(NV) in the sinusoids or internalized in the hepatic cells.


Assuntos
Materiais Biocompatíveis/farmacologia , Fígado/metabolismo , Nanodiamantes/química , Animais , Células Hep G2 , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Imageamento Tridimensional , Cinética , Fígado/efeitos dos fármacos , Microscopia de Fluorescência , Tamanho da Partícula , Ratos Sprague-Dawley , Distribuição Tecidual
4.
Brain Res ; 1234: 158-71, 2008 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-18708033

RESUMO

Deposition of beta-amyloid (Abeta) peptides in the walls of brain blood vessels, cerebral amyloid angiopathy (CAA), is common in patients with Alzheimer's disease (AD). Previous studies have demonstrated Abeta peptide deposition among vascular smooth muscle cells (VSMCs), but the source of the Abeta and basis for its selective deposition in VSMCs are unknown. In the present study, we examined the deposition patterns of Abeta peptides, Abeta40 and Abeta42, within the cerebrovasculature of AD and control patients using single- and double-label immunohistochemistry. Abeta40 and Abeta42 were abundant in VSMCs, especially in leptomeningeal arteries and their initial cortical branches; in later-stage AD brains this pattern extended into the microvasculature. Abeta peptide deposition was linked to loss of VSMC viability. Perivascular leak clouds of Abeta-positive material were associated primarily with arterioles. By contrast, control brains possessed far fewer Abeta42- and Abeta40-immunopositive blood vessels, with perivascular leak clouds of Abeta-immunopositive material rarely observed. We also demonstrate that VSMCs in brain blood vessels express the alpha7 nicotinic acetylcholine receptor (alpha7nAChR), which has high binding affinity for Abeta peptides, especially Abeta42. These results suggest that the blood and blood-brain barrier permeability provide a major source of the Abeta peptides that gradually deposit in brain VSMCs, and the presence and abundance of the alpha7nAChR on VSMCs may facilitate the selective accumulation of Abeta peptides in these cells.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Angiopatia Amiloide Cerebral/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Receptores Nicotínicos/biossíntese , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Arteríolas/metabolismo , Arteríolas/patologia , Barreira Hematoencefálica/fisiologia , Sobrevivência Celular , Angiopatia Amiloide Cerebral/patologia , Artérias Cerebrais/metabolismo , Artérias Cerebrais/patologia , Córtex Entorrinal/patologia , Feminino , Hipocampo/patologia , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Músculo Liso Vascular/citologia , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Receptor Nicotínico de Acetilcolina alfa7
5.
J Comput Biol ; 14(3): 350-9, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17563316

RESUMO

MOTIVATION: DNA microarrays are a well-known and established technology in biological and pharmaceutical research providing a wealth of information essential for understanding biological processes and aiding drug development. Protein microarrays are quickly emerging as a follow-up technology, which will also begin to experience rapid growth as the challenges in protein to spot methodologies are overcome. Like DNA microarrays, their protein counterparts produce large amounts of data that must be suitably analyzed in order to yield meaningful information that should eventually lead to novel drug targets and biomarkers. Although the statistical management of DNA microarray data has been well described, there is no available report that offers a successful consolidated approach to the analysis of high-throughput protein microarray data. We describe the novel application of a statistical methodology to analyze the data from an immune response profiling assay using human protein microarray with over 5000 proteins on each chip.


Assuntos
Análise Serial de Proteínas/métodos , Autoanticorpos/sangue , Doenças Autoimunes/sangue , Doenças Autoimunes/imunologia , Interpretação Estatística de Dados , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Software
6.
Brain Res ; 1142: 223-36, 2007 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-17306234

RESUMO

We have investigated the possibility that soluble, blood-borne amyloid beta (Abeta) peptides can cross a defective blood-brain barrier (BBB) and interact with neurons in the brain. Immunohistochemical analyses revealed extravasated plasma components, including Abeta42 in 19 of 21 AD brains, but in only 3 of 13 age-matched control brains, suggesting that a defective BBB is common in AD. To more directly test whether blood-borne Abeta peptides can cross a defective BBB, we tracked the fate of fluorescein isothiocyanate (FITC)-labeled Abeta42 and Abeta40 introduced via tail vein injection into mice with a BBB rendered permeable by treatment with pertussis toxin. Both Abeta40 and Abeta42 readily crossed the permeabilized BBB and bound selectively to certain neuronal subtypes, but not glial cells. By 48 h post-injection, Abeta42-positive neurons were widespread in the brain. In the cerebral cortex, small fluorescent, Abeta42-positive granules were found in the perinuclear cytoplasm of pyramidal neurons, suggesting that these cells can internalize exogenous Abeta42. An intact BBB (saline-injected controls) blocked entry of blood-borne Abeta peptides into the brain. The neuronal subtype selectivity of Abeta42 and Abeta40 was most evident in mouse brains subjected to direct intracranial stereotaxic injection into the hippocampal region, thereby bypassing the BBB. Abeta40 was found to preferentially bind to a distinct subset of neurons positioned at the inner face of the dentate gyrus, whereas Abeta42 bound selectively to the population of large neurons in the hilus region of the dentate gyrus. Our results suggest that the blood may serve as a major, chronic source of soluble, exogenous Abeta peptides that can bind selectively to certain subtypes of neurons and accumulate within these cells.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Barreira Hematoencefálica/fisiopatologia , Encéfalo/patologia , Permeabilidade Capilar/fisiologia , Neurônios/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Permeabilidade Capilar/efeitos dos fármacos , Estudos de Casos e Controles , Colágeno Tipo IV/metabolismo , Imunofluorescência/métodos , Humanos , Indóis , Camundongos , Toxina Pertussis/farmacologia , Mudanças Depois da Morte , Transporte Proteico/fisiologia , Fatores de Tempo
7.
BMC Physiol ; 7: 3, 2007 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-17397547

RESUMO

BACKGROUND: Protease-activated receptors (PAR) are present in the urinary bladder, and their expression is altered in response to inflammation. PARs are a unique class of G protein-coupled that carry their own ligands, which remain cryptic until unmasked by proteolytic cleavage. Although the canonical signal transduction pathway downstream of PAR activation and coupling with various G proteins is known and leads to the rapid transcription of genes involved in inflammation, the effect of PAR activation on the downstream transcriptome is unknown. We have shown that intravesical administration of PAR-activating peptides leads to an inflammatory reaction characterized by edema and granulocyte infiltration. Moreover, the inflammatory response to intravesical instillation of known pro-inflammatory stimuli such as E. coli lipopolysaccharide (LPS), substance P (SP), and antigen was strongly attenuated by PAR1- and to a lesser extent by PAR2-deficiency. RESULTS: Here, cDNA array experiments determined inflammatory genes whose expression is dependent on PAR1 activation. For this purpose, we compared the alteration in gene expression in wild type and PAR1-/- mice induced by classical pro-inflammatory stimuli (LPS, SP, and antigen). 75 transcripts were considered to be dependent on PAR-1 activation and further annotated in silico by Ingenuity Pathways Analysis (IPA) and gene ontology (GO). Selected transcripts were target validated by quantitative PCR (Q-PCR). Among PAR1-dependent transcripts, the following have been implicated in the inflammatory process: b2m, ccl7, cd200, cd63, cdbpd, cfl1, dusp1, fkbp1a, fth1, hspb1, marcksl1, mmp2, myo5a, nfkbia, pax1, plaur, ppia, ptpn1, ptprcap, s100a10, sim2, and tnfaip2. However, a balanced response to signals of injury requires a transient cellular activation of a panel of genes together with inhibitory systems that temper the overwhelming inflammation. In this context, the activation of genes such as dusp1 and nfkbia seems to counter-balance the inflammatory response to PAR activation by limiting prolonged activation of p38 MAPK and increased cytokine production. In contrast, transcripts such as arf6 and dcnt1 that are involved in the mechanism of PAR re-sensitization would tend to perpetuate the inflammatory reaction in response to common pro-inflammatory stimuli. CONCLUSION: The combination of cDNA array results and genomic networks reveals an overriding participation of PAR1 in bladder inflammation, provides a working model for the involvement of downstream signaling, and evokes testable hypotheses regarding the transcriptome downstream of PAR1 activation. It remains to be determined whether or not mechanisms targeting PAR1 gene silencing or PAR1 blockade will ameliorate the clinical manifestation of cystitis.


Assuntos
Cistite/genética , Cistite/metabolismo , Regulação da Expressão Gênica , Receptor PAR-1/metabolismo , Animais , Antígenos/imunologia , Cálcio/metabolismo , Cromatina/metabolismo , Cistite/induzido quimicamente , Cistite/imunologia , Feminino , Expressão Gênica , Genoma , Imunoprecipitação , Lipopolissacarídeos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Fosfolipases A/metabolismo , Reação em Cadeia da Polimerase/métodos , Receptor PAR-1/deficiência , Frações Subcelulares/metabolismo , Substância P , Bexiga Urinária/metabolismo
8.
BMC Physiol ; 7: 4, 2007 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-17397548

RESUMO

BACKGROUND: In general, inflammation plays a role in most bladder pathologies and represents a defense reaction to injury that often times is two edged. In particular, bladder neurogenic inflammation involves the participation of mast cells and sensory nerves. Increased mast cell numbers and tryptase release represent one of the prevalent etiologic theories for interstitial cystitis and other urinary bladder inflammatory conditions. The activity of mast cell-derived tryptase as well as thrombin is significantly increased during inflammation. Those enzymes activate specific G-protein coupled proteinase-activated receptors (PAR)s. Four PARs have been cloned so far, and not only are all four receptors highly expressed in different cell types of the mouse urinary bladder, but their expression is altered during experimental bladder inflammation. We hypothesize that PARs may link mast cell-derived proteases to bladder inflammation and, therefore, play a fundamental role in the pathogenesis of cystitis. RESULTS: Here, we demonstrate that in addition to the mouse urinary bladder, all four PA receptors are also expressed in the J82 human urothelial cell line. Intravesical administration of PAR-activating peptides in mice leads to an inflammatory reaction characterized by edema and granulocyte infiltration. Moreover, the inflammatory response to intravesical instillation of known pro-inflammatory stimuli such as E. coli lipopolysaccharide (LPS), substance P, and antigen was strongly attenuated by PAR1-, and to a lesser extent, by PAR2-deficiency. CONCLUSION: Our results reveal an overriding participation of PAR1 in bladder inflammation, provide a working model for the involvement of downstream signaling, and evoke testable hypotheses regarding the role of PARs in bladder inflammation. It remains to be determined whether or not mechanisms targeting PAR1 gene silencing or PAR1 blockade will ameliorate the clinical manifestations of cystitis.


Assuntos
Cistite/metabolismo , Receptor PAR-1/metabolismo , Animais , Antígenos/imunologia , Linhagem Celular , Cistite/induzido quimicamente , Cistite/imunologia , Cistite/patologia , Edema/induzido quimicamente , Granulócitos/patologia , Humanos , Lipopolissacarídeos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor PAR-1/agonistas , Receptor PAR-1/deficiência , Receptor PAR-2/efeitos dos fármacos , Receptor PAR-2/metabolismo , Receptores Ativados por Proteinase/metabolismo , Substância P , Bexiga Urinária/metabolismo , Urotélio/citologia , Urotélio/metabolismo
9.
Trends Neurosci ; 26(9): 496-500, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12948661

RESUMO

The discovery of proteinase-activated receptors (PARs) in the nervous system has led to new insights about the potential physiological functions of these enzymes, which were traditionally considered merely as degradative molecules. This review summarizes evidence that proteinases, through activation of PARs, interact with the peripheral nervous system (PNS), playing roles in neurogenic inflammation, pain perception, secretory and motor functions, as well as in the response to nerve injuries. Activation of PARs interferes with numerous physiological events that are under tight neural control, in addition to modulating nerve survival. New potential roles are suggested for members of the PAR family, highlighting proteinases and their receptors as potential therapeutic targets for diseases associated with PNS activation.


Assuntos
Endopeptidases/metabolismo , Neurônios/metabolismo , Sistema Nervoso Periférico/metabolismo , Receptores de Trombina/metabolismo , Animais , Secreções Corporais/metabolismo , Sobrevivência Celular , Endopeptidases/química , Inflamação/metabolismo , Movimento , Regeneração Nervosa , Dor/metabolismo , Receptor PAR-1 , Transdução de Sinais
10.
Curr Pharm Des ; 12(6): 677-84, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16472157

RESUMO

Although there is still no known effective preventative treatment or cure for Alzheimer's disease (AD), the development of new drugs that target pathological features that appear early in the course of this disease and alleviate some of the early cognitive and memory symptoms is a laudable goal that may be one step closer. To date, the acetylcholinesterase inhibitors have been the most widely used AD drugs and have been somewhat successful in slowing loss of cognition. In the last few years, a number of studies have demonstrated that amyloid beta (1-42) (Abeta42), the predominant Abeta peptide species in amyloid plaques, first accumulates in vulnerable neurons prior to plaque formation. Recently, we have shown that many (if not most) amyloid plaques in the entorhinal cortex of AD brains are actually the lysis remnants of degenerated, Abeta42-overburdened neurons. Furthermore, the most vulnerable neurons appear to be those that abundantly express the alpha7 nicotinic acetylcholine receptor (alpha7nAChR), and internalization of Abeta42 appears to be facilitated by the high-affinity binding of Abeta42 to the alpha7nAChR on neuronal cell surfaces, followed by endocytosis of the resulting complex and its accumulation within the lysosomal compartment. This mechanism provides a reasonable explanation for the selective vulnerability of cholinergic and cholinoceptive neurons in AD brains and for the fact that Abeta42 is the dominant Abeta peptide species in both intraneuronal accumulations and amyloid plaques. In view of the pathophysiological consequences of Abeta42 binding to alpha7nAChR on neuronal surfaces that stem from excessive intraneuronal Abeta42 accumulation, the alpha7nAChR could be an important therapeutic target for treatment of AD. In addition, it further emphasizes the potential merits of new and effective therapeutic strategies pointed towards the goal of lowering of Abeta42 levels in the blood and cerebrospinal fluid as well as blocking Abeta42 in the blood from penetrating the blood-brain barrier and entering into the brain parenchyma.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/metabolismo , Antagonistas Nicotínicos/uso terapêutico , Fragmentos de Peptídeos/metabolismo , Células Piramidais/efeitos dos fármacos , Receptores Nicotínicos/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Microscopia Eletrônica , Antagonistas Nicotínicos/farmacologia , Placa Amiloide/metabolismo , Placa Amiloide/ultraestrutura , Células Piramidais/metabolismo , Células Piramidais/patologia , Receptor Nicotínico de Acetilcolina alfa7
11.
Mol Cancer Ther ; 4(8): 1198-204, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16093435

RESUMO

With the advent of agents directed against specific molecular targets in drug discovery, it has become imperative to show a compound's cellular impact on the intended biomolecule in vivo. The objective of the present study was to determine if we could develop an assay to validate the in vivo effects of a compound. Hence, we investigated the in vivo pharmacodynamic activity of JNJ-10198409, a relatively selective inhibitor of platelet-derived growth factor receptor tyrosine kinase (PDGF-RTK), in tumor tissues after administering the compound orally in a nude mouse xenograft model of human LoVo colon cancer. We developed a novel assay to quantify the in vivo anti-PDGF-RTK activity of the inhibitor in tumor tissue by determining the phosphorylation status of phospholipase Cgamma1 (PLCgamma1), a key downstream cellular molecule in the PDGF-RTK signaling cascade. We used two antibodies, one specific for the total (phosphorylated and unphosphorylated forms) PLCgamma1 (pan-PLCgamma1) and the other, specific for phosphorylated form of PLCgamma1 (ph-PLCgamma1) to immunohistochemically detect their expression in tumor tissues. Computer-assisted image analysis was then used to directly compare the ratio of ph-PLCgamma1 to pan-PLCgamma1 immunolabeling intensities in serial sections (5 mum) of tumors obtained from vehicle- and JNJ-10198409-treated tumor-bearing mice. Our data showed statistically significant, dose-dependent differences in the ph-PLC/pan-PLC ratio among the four treatment groups (vehicle, 25, 50, and 100 mg/kg b.i.d.). These results confirmed this compound's ability to suppress PDGF-RTK downstream signaling in tumor tissues in vivo. In addition to this specific application of this in vivo validation approach to those targets that use PLCgamma as a downstream signaling partner, these methods may also benefit other drug discovery targets.


Assuntos
Antineoplásicos/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Indanos/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Pirazóis/farmacologia , Receptores do Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Animais , Anticorpos Fosfo-Específicos/imunologia , Antineoplásicos/química , Neoplasias do Colo/enzimologia , Humanos , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Indanos/química , Camundongos , Fosfolipase C gama , Fosforilação , Inibidores de Proteínas Quinases/química , Pirazóis/química , Fosfolipases Tipo C/análise , Fosfolipases Tipo C/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Endocrinology ; 146(3): 1066-73, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15564322

RESUMO

Deleted in malignant brain tumors 1 (DMBT1) is a candidate suppressor of malignancies of the brain, lung, gut, and breast. We have been studying gene expression in the uterus in the presence of estrogens and their antagonists. Here, we show that DMBT1 RNA levels are robustly increased by estrogen treatment in the uteri of ovariectomized monkeys and rats. In monkeys, the progestin antagonist mifepristone inhibits estrogen-dependent uterine proliferation. As determined by a microarray experiment and quantitative analysis of RNA levels, mifepristone inhibited estrogenic induction of DMBT1. DMBT1 was not expressed in intact monkeys that were treated with a gonadotropin agonist to suppress steroidogenesis. An in vitro transfection study with human DMBT1 promoter constructs showed that an Alu site approximately 3000 nucleotides upstream of the gene mediates estrogenic regulation. Surprisingly, the estrogen antagonists tamoxifen, raloxifene, and ICI 182,780 also induced gene expression via this Alu site. Rodents represent a more convenient model system for studying uterine biology than monkeys. In rats, uterine DMBT1 RNA levels were dramatically up-regulated by estrogen. Consistent with the transfection study, tamoxifen and raloxifene increased DMBT1 RNA levels in vivo, but ICI 182,780 inhibited an estrogen-induced increase. Immunohistochemical studies showed that DMBT1 is specifically induced in glandular and luminal epithelia of the rat endometrium. Our experiments establish that DMBT1 is an estrogen-responsive gene with a possible role in endometrial proliferation or differentiation, and they have implications for the putative tumor suppressive and mucosal protective functions of DMBT1 in the uterus.


Assuntos
Aglutininas/fisiologia , Endométrio/metabolismo , Epitélio/metabolismo , Estradiol/análogos & derivados , Estrogênios/metabolismo , Regulação da Expressão Gênica , Mucinas/fisiologia , Receptores de Superfície Celular/fisiologia , Elementos Alu , Animais , Northern Blotting , Proteínas de Ligação ao Cálcio , Diferenciação Celular , Linhagem Celular , Proteínas de Ligação a DNA , Relação Dose-Resposta a Droga , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Feminino , Fulvestranto , Haplorrinos , Humanos , Imuno-Histoquímica , Luciferases/metabolismo , Mucinas/biossíntese , Mucosa/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Plasmídeos/metabolismo , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , RNA/química , RNA/metabolismo , Cloridrato de Raloxifeno/farmacologia , Ratos , Ratos Sprague-Dawley , Tamoxifeno/farmacologia , Transfecção , Proteínas Supressoras de Tumor , Regulação para Cima , Útero/metabolismo
13.
Curr Top Med Chem ; 5(11): 1047-51, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16181130

RESUMO

Biomarkers in the clinical oncology field can have tremendous therapeutic impact especially if the marker is detected before clinical symptoms. This impact can be extended to the evaluation of clinical oncology treatments allowing evaluation of potential compounds to determine their efficacy in the disease treatment. The discovery of clinical biomarkers can consume time, resources and costs. Therefore, it is important that the most effective strategies are employed to discover these biomarkers. These strategies may include the integration of available genomic, proteomic and histopathological technologies, which could reduce the costs and aid in the validation of the biomarker. Certainly the type of biomarker needed to address a particularly defined problem will drive the type of technology. However, a single biomarker to diagnose a specific cancer can be as elusive as relying on a single technology. This review examines some of the technologies used to discover biomarkers and presents the use of combinatorial technical synergies to discover and validate potential clinical oncology biomarkers.


Assuntos
Biomarcadores Tumorais , Avaliação Pré-Clínica de Medicamentos/métodos , Genômica/métodos , Histocitoquímica/métodos , Humanos , Proteômica/métodos
14.
J Histochem Cytochem ; 53(7): 895-903, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15995148

RESUMO

Cyclic nucleotide phosphodiesterase 11A (PDE11A) is the newest member in the PDE family. Although the tissue distribution of PDE11A mRNA has been shown, its protein expression pattern has not been well studied. The goal of this report is to investigate the distribution of PDE11A proteins in a wide range of normal and malignant human tissues. We utilized a polyclonal antibody that recognized all four PDE11A isoforms. Its specificity was demonstrated by Western blot analysis on a recombinant human PDE11A protein and native PDE11A proteins in various human tissues. Immunohistochemistry showed that PDE11A is widely expressed. Various degrees of immunoreactivity were observed in the epithelial cells, endothelial cells, and smooth muscle cells of all tissues examined. The highest expression was in the epithelial, endothelial, and smooth muscle cells of the prostate, Leydig, and spermatogenic cells of the testis, the tubule epithelial cells in the kidney, the epithelial and endothelial cells in the adrenal, the epithelial cells and macrophages in the colon, and the epidermis in the skin. Furthermore, PDE11A expression was also detected in several human carcinomas. Our results suggest that PDE11A might be involved in multiple physiological processes in various organs via its ability to modulate intracellular cAMP and cGMP levels.


Assuntos
Neoplasias/enzimologia , Diester Fosfórico Hidrolases/biossíntese , 3',5'-GMP Cíclico Fosfodiesterases , Células Endoteliais/enzimologia , Células Epiteliais/enzimologia , Humanos , Imuno-Histoquímica , Miócitos de Músculo Liso/enzimologia , Especificidade de Órgãos
15.
J Mol Neurosci ; 25(3): 225-38, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15800376

RESUMO

Activity-dependent neurotrophic factor (ADNF) is a novel, femtomolar-acting, glial-derived polypeptide (14 kDa) known to protect neurons from a variety of toxic insults. The active site for ADNF function is localized to a 9-amino-acid stretch (SALLRSIPA; ADNF-9). A few years later, a novel ADNF-9-like active peptide (NAPVSIPQ or NAP) was identified and shown to be expressed in the CNS and exhibit an activity profile similar to ADNF-9. Such studies suggest that ADNF-9 and NAP might function like other known neurotrophins and play a role in neural development and maintenance. The purpose of the present studies was to determine if ADNF-9 or NAP affects neurite outgrowth and synaptogenesis in rat hippocampal and cortical cultures. Using MAP2-FITC immunofluorescent labeling, we found that ADNF-9 and NAP promoted neurite outgrowth in a concentration-dependent manner, with maximal activity observed at femtomolar concentrations. Both peptides stimulated robust outgrowth in hippocampal cells (approximately 150% of control; p < 0.01) with a modest effect on cortical cells (approximately 20% of control; p < 0.05) similar to other known growth factors. However, the outgrowth-promoting effect was abolished in the absence of serum, suggesting that soluble factors might be necessary for the neurotrophic activity. Finally, we found that ADNF-9 and NAP increased synaptophysin expression in both rat hippocampal and cortical cultures. These results suggest that ADNF-9 and NAP might contribute to neuronal plasticity associated with development and repair after injury.


Assuntos
Proteínas do Tecido Nervoso/farmacologia , Neuritos/efeitos dos fármacos , Neuritos/fisiologia , Neurônios/fisiologia , Oligopeptídeos/farmacologia , Animais , Células Cultivadas , Córtex Cerebral/citologia , Feminino , Hipocampo/citologia , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Neurônios/ultraestrutura , Gravidez , Ratos , Sinaptofisina/metabolismo
16.
Med Hypotheses ; 64(3): 458-63, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15617848

RESUMO

A sole pathological event leading to Alzheimer's disease (AD) remains undiscovered in spite of decades of costly research. In fact, it is more probable that the causes of AD are the result of a myriad of intertwining pathologies. However, hope remains that a single awry event could lead to the many pathological events observed in AD brain tissues thereby creating the presentation of simultaneous pathologies. Age-related vascular diseases, which include an impaired blood-brain barrier (BBB), are a common denominator associated with various degrees of dementia, including AD. Recently, a key finding not only demonstrated the anomalous presence of immunoglobulin (Ig) detection in the brain parenchyma of AD tissues but, most importantly, specific neurons that showed degenerative, apoptotic features contained these vascular-derived antibodies. In addition, subsequent studies detected classical complement components, C1q and C5b-9, in these Ig-positive neurons, which also were spatially more associated with reactive microglia over the Ig-negative neurons. Thus, it is possible that the mere presence of anti-neuronal autoantibodies in the serum, whose importance had been previously dismissed, may be without pathological consequence until there is a BBB dysfunction to allow the deleterious effects of these autoantibodies access on their targets. Hence, these observations suggest autoimmunity-induced cell death in AD.


Assuntos
Doença de Alzheimer/imunologia , Doenças Autoimunes , Autoimunidade , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Autoanticorpos/sangue , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Complemento C1q/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento , Proteínas do Sistema Complemento/metabolismo , Glicoproteínas/metabolismo , Humanos , Imunoglobulina G/análise , Neurônios/metabolismo
17.
Am J Alzheimers Dis Other Demen ; 20(3): 144-50, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16003929

RESUMO

A recent study provided evidence that immunoglobulins (Igs) are not only present in Alzheimer's disease (AD) brains, but are also immunohistochemically detected in and/or on a particular population of pyramidal neurons that appeared morphologically degenerative in contrast to neighboring normal-appearing Ig-negative neurons. Because little has been reported about these Ig-positive neurons, the objectives of this study were to characterize the inflammatory profile of these neurons in the AD brain by determining if they possess complement components and are associated with reactive microglia. The data showed that the Ig-positive neurons had complement C1q and C5b-9 proteins and appeared degenerative. Furthermore, D-related human leukocyte antigen (HLA-DR)-positive fibers of reactive microglia were spatially closer (p < 0.001) and often in contact with the Ig-positive neurons than the Ig-negative neurons. Collectively, these data suggest that the Ig-positive neurons detected in AD brains are dying from the processes of the antibody-induced classical complement pathway.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Complemento C1q/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Antígenos HLA-DR/metabolismo , Imunoglobulinas/metabolismo , Vias Neurais/metabolismo , Vias Neurais/patologia , Células Piramidais/metabolismo , Células Piramidais/patologia , Córtex Entorrinal/metabolismo , Córtex Entorrinal/patologia , Humanos , Imuno-Histoquímica
18.
Neurobiol Aging ; 25(5): 675-83, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15172747

RESUMO

Alzheimer disease (AD) involves glial inflammation associated with amyloid plaques. The role of the microglial cells in the AD brain is controversial, as it remains unclear if the microglia form the amyloid fibrils of plaques or react to them in a macrophage-phagocytic role. Also, it is not known why microglia are preferentially associated with some amyloid plaque types. This review will provide substantial evidence to support the phagocytic role of microglia in the brain as well as explain why microglia are generally associated with specific plaque types that may be explained through their unique mechanisms of formation. In summary, the data presented suggests that plaque associated microglial activation is typically subsequent to specific amyloid plaque formations in the AD brain.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/patologia , Microglia/patologia , Fagocitose/fisiologia , Placa Amiloide/metabolismo , Doença de Alzheimer/metabolismo , Animais , Encéfalo/ultraestrutura , Humanos , Modelos Biológicos
19.
Neurobiol Aging ; 23(2): 213-23, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-11804705

RESUMO

beta-amyloid(1-42) (Abeta42)-rich amyloid plaques (APs) may be derived from destroyed neurons that were burdened with extensive intracellular Abeta42 accumulations. Since most cells that accumulate Abeta42 express the alpha7 nicotinic acetylcholine receptor (alpha7nAChR), we examined the relationship between the intracellular accumulation of Abeta42 and the expression of the alpha7nAChR in cells from the cerebellum of sporadic Alzheimer's disease (AD) patients. Abeta42, but not Abeta40 or Abeta43, accumulates intracellularly in Purkinje, Golgi II, stellate and basket cells in the AD cerebellum, all of which express the alpha7nAChR. Abeta42 deposits were also prominent within dendrites of Purkinje cells, especially at points of their bifurcation that were often occluded with this material. Diffuse APs appeared to represent the remnants of destroyed Abeta42-laden segments of Purkinje cell dendritic trees. Similarly, the accumulation of Abeta42 and early loss of Golgi II cells in AD cerebella correlated directly to their high level of alpha7nAChR expression. Furthermore, the presence and relative abundance of neuron-derived Abeta42/alpha7nAChR-positive materials within Bergman glia may be indicative of the stage of AD. These data are consistent with a role for the alpha7nAChR in mediating intracellular Abeta42 accumulation and also support the notion that the intracellular and intradendritic accumulation of Abeta42 may eventually result in cell lysis and the formation of APs.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Cerebelo/metabolismo , Cerebelo/patologia , Dendritos/metabolismo , Dendritos/patologia , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Células de Purkinje/metabolismo , Células de Purkinje/patologia , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Imuno-Histoquímica , Masculino , Neurônios/metabolismo , Receptores Nicotínicos/metabolismo , Receptor Nicotínico de Acetilcolina alfa7
20.
J Histochem Cytochem ; 51(5): 687-96, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12704216

RESUMO

Sphingolipids serve as structural elements of cells and as lipid second messengers. They regulate cellular homeostasis, mitogenesis, and apoptosis. Sphingolipid signaling may also be important in various pathophysiologies such as vascular injury, inflammation, and cancer. Serine palmitoyltransferase (SPT) catalyzes the condensation of serine with palmitoyl-CoA, the first, rate-limiting step in de novo sphingolipid biosynthesis. This integral microsomal membrane protein consists of at least two subunits, SPT1 and SPT2. In this study we analyzed the expression of SPT1 and SPT2 in normal human tissues. Strong SPT1 and SPT2 expression was observed in pyramidal neurons in the brain, in colon epithelium, and in mucosal macrophages. However, SPT2 expression was more prominent than SPT1 in the colon mucosal macrophages, the adrenomedullary chromaffin cells and endothelium, and in the uterine endothelium. SPT2 was localized in both nuclei and cytoplasm of the adrenomedullary chromaffin cells, whereas SPT1 was primarily cytoplasmic. These observations link enhanced SPT expression to proliferating cells, such as the lung, stomach, intestinal epithelium, and renal proximal tubular epithelium, and to potentially activated cells such as neurons, chromaffin cells, and mucosal macrophages. A baseline expression of SPT, established by this study, may serve as a measure for aberrant expression in various disease states.


Assuntos
Aciltransferases/metabolismo , Aciltransferases/imunologia , Especificidade de Anticorpos , Humanos , Imuno-Histoquímica , Especificidade de Órgãos , Subunidades Proteicas , Serina C-Palmitoiltransferase
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa