Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Toxicol Pathol ; 51(7-8): 470-481, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38288963

RESUMO

Toxicogenomic technologies query the genome, transcriptome, proteome, and the epigenome in a variety of toxicological conditions. Due to practical considerations related to the dynamic range of the assays, sensitivity, cost, and technological limitations, transcriptomic approaches are predominantly used in toxicogenomics. Toxicogenomics is being used to understand the mechanisms of toxicity and carcinogenicity, evaluate the translational relevance of toxicological responses from in vivo and in vitro models, and identify predictive biomarkers of disease and exposure. In this session, a brief overview of various transcriptomic technologies and practical considerations related to experimental design was provided. The advantages of gene network analyses to define mechanisms were also discussed. An assessment of the utility of toxicogenomic technologies in the environmental and pharmaceutical space showed that these technologies are being increasingly used to gain mechanistic insights and determining the translational relevance of adverse findings. Within the environmental toxicology area, there is a broader regulatory consideration of benchmark doses derived from toxicogenomics data. In contrast, these approaches are mainly used for internal decision-making in pharmaceutical development. Finally, the development and application of toxicogenomic signatures for prediction of apical endpoints of regulatory concern continues to be area of intense research.


Assuntos
Fígado , Toxicogenética , Proteômica , Perfilação da Expressão Gênica , Transcriptoma
2.
Int J Mol Sci ; 22(23)2021 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-34884436

RESUMO

Cancer therapies targeting the vascular endothelial growth factor (VEGF) signaling pathway can lead to renal damage by disrupting the glomerular ultrafiltration apparatus. The objective of the current study was to identify sensitive biomarkers for VEGF inhibition-induced glomerular changes in rats. Male Sprague-Dawley rats were administered an experimental VEGF receptor (VEGFR) inhibitor, ABT-123, for seven days to investigate the correlation of several biomarkers with microscopic and ultrastructural changes. Glomeruli obtained by laser capture microdissection were also subjected to gene expression analysis to investigate the underlying molecular events of VEGFR inhibition in glomerulus. ABT-123 induced characteristic glomerular ultrastructural changes in rats, including fusion of podocyte foot processes, the presence of subendothelial electron-dense deposits, and swelling and loss of fenestrations in glomerular endothelium. The subtle morphological changes cannot be detected with light microscopy or by changes in standard clinical chemistry and urinalysis. However, urinary albumin increased 44-fold as early as Day three. Urinary ß2-microglobulin levels were also increased. Other urinary biomarkers that are typically associated with tubular injury were not significantly impacted. Such patterns in urinary biomarkers can provide valuable diagnostic insight to VEGF inhibition therapy-induced glomeruli injuries.


Assuntos
Nefropatias/urina , Inibidores de Proteínas Quinases/efeitos adversos , Transdução de Sinais/efeitos dos fármacos , Microglobulina beta-2/urina , Albuminas/metabolismo , Animais , Biomarcadores/urina , Modelos Animais de Doenças , Nefropatias/induzido quimicamente , Nefropatias/metabolismo , Microdissecção e Captura a Laser , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
Front Big Data ; 2: 25, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-33693348

RESUMO

Most small molecule drugs interact with unintended, often unknown, biological targets and these off-target interactions may lead to both preclinical and clinical toxic events. Undesired off-target interactions are often not detected using current drug discovery assays, such as experimental polypharmacological screens. Thus, improvement in the early identification of off-target interactions represents an opportunity to reduce safety-related attrition rates during preclinical and clinical development. In order to better identify potential off-target interactions that could be linked to predictable safety issues, a novel computational approach to predict safety-relevant interactions currently not covered was designed and evaluated. These analyses, termed Off-Target Safety Assessment (OTSA), cover more than 7,000 targets (~35% of the proteome) and > 2,46,704 preclinical and clinical alerts (as of January 20, 2019). The approach described herein exploits a highly curated training set of >1 million compounds (tracking >20 million compound-structure activity relationship/SAR data points) with known in vitro activities derived from patents, journals, and publicly available databases. This computational process was used to predict both the primary and secondary pharmacological activities for a selection of 857 diverse small molecule drugs for which extensive secondary pharmacology data are readily available (456 discontinued and 401 FDA approved). The OTSA process predicted a total of 7,990 interactions for these 857 molecules. Of these, 3,923 and 4,067 possible high-scoring interactions were predicted for the discontinued and approved drugs, respectively, translating to an average of 9.3 interactions per drug. The OTSA process correctly identified the known pharmacological targets for >70% of these drugs, but also predicted a significant number of off-targets that may provide additional insight into observed in vivo effects. About 51.5% (2,025) and 22% (900) of these predicted high-scoring interactions have not previously been reported for the discontinued and approved drugs, respectively, and these may have a potential for repurposing efforts. Moreover, for both drug categories, higher promiscuity was observed for compounds with a MW range of 300 to 500, TPSA of ~200, and clogP ≥7. This computation also revealed significantly lower promiscuity (i.e., number of confirmed off-targets) for compounds with MW > 700 and MW<200 for both categories. In addition, 15 internal small molecules with known off-target interactions were evaluated. For these compounds, the OTSA framework not only captured about 56.8% of in vitro confirmed off-target interactions, but also identified the right pharmacological targets for 14 compounds as one of the top scoring targets. In conclusion, the OTSA process demonstrates good predictive performance characteristics and represents an additional tool with utility during the lead optimization stage of the drug discovery process. Additionally, the computed physiochemical properties such as clogP (i.e., lipophilicity), molecular weight, pKa and logS (i.e., solubility) were found to be statistically different between the approved and discontinued drugs, but the internal compounds were close to the approved drugs space in most part.

6.
Int J Mol Sci ; 11(11): 4697-714, 2010 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-21151465

RESUMO

Idiosyncratic drug reactions (IDRs) are poorly understood, unpredictable, and not detected in preclinical studies. Although the cause of these reactions is likely multi-factorial, one hypothesis is that an underlying inflammatory state lowers the tolerance to a xenobiotic. Previously used in an inflammation IDR model, bacterial lipopolysaccharide (LPS) is heterogeneous in nature, making development of standardized testing protocols difficult. Here, the use of rat tumor necrosis factor-α (TNFα) to replace LPS as an inflammatory stimulus was investigated. Sprague-Dawley rats were treated with separate preparations of LPS or TNFα, and hepatic transcriptomic effects were compared. TNFα showed enhanced consistency at the transcriptomic level compared to LPS. TNFα and LPS regulated similar biochemical pathways, although LPS was associated with more robust inflammatory signaling than TNFα. Rats were then codosed with TNFα and trovafloxacin (TVX), an IDR-associated drug, and evaluated by liver histopathology, clinical chemistry, and gene expression analysis. TNFα/TVX induced unique gene expression changes that clustered separately from TNFα/levofloxacin, a drug not associated with IDRs. TNFα/TVX cotreatment led to autoinduction of TNFα resulting in potentiation of underlying gene expression stress signals. Comparison of TNFα/TVX and LPS/TVX gene expression profiles revealed similarities in the regulation of biochemical pathways. In conclusion, TNFα could be used in lieu of LPS as an inflammatory stimulus in this model of IDRs.


Assuntos
Anti-Infecciosos/toxicidade , Fluoroquinolonas/toxicidade , Lipopolissacarídeos/toxicidade , Fígado/efeitos dos fármacos , Naftiridinas/toxicidade , Fator de Necrose Tumoral alfa/toxicidade , Animais , Anti-Infecciosos/antagonistas & inibidores , Interações Medicamentosas , Fluoroquinolonas/antagonistas & inibidores , Inflamação/induzido quimicamente , Inflamação/metabolismo , Lipopolissacarídeos/antagonistas & inibidores , Fígado/metabolismo , Naftiridinas/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Transcriptoma , Fator de Necrose Tumoral alfa/antagonistas & inibidores
8.
J Invest Surg ; 19(4): 245-54, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16835139

RESUMO

Disturbances of angiogenesis have been suggested to result in the impaired healing of skin wounds. Using a murine incisional wound model, we evaluated the effects of SU6668, an inhibitor of the receptors for vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), and fibroblast growth factor (FGF), on the healing of skin wounds. Mice were administered vehicle, SU6668 (100 or 400 mg/kg/day, b.i.d.), or dexamethasone (1 mg/kg/day, b.i.d.), and wound healing was monitored histologically and using a tensiometer. SU6668 at a fully efficacious dose of 100 mg/kg/day had no significant effect on the healing process, while at a supratherapeutic dose of 400 mg/kg/day, there were subtle transient histologic changes and slight decreases in tensile strength, suggesting a slight delay in the wound healing process. In conclusion, these data indicate that inhibition of the receptors for VEGF, PDGF, and FGF at levels necessary to inhibit tumor growth in mouse xenograft models does not affect the healing of incisional wounds in mice. Redundant pathways likely compensate for inhibition of VEGF, PDGF, and FGF signaling pathways in the skin healing process.


Assuntos
Indóis/uso terapêutico , Pirróis/uso terapêutico , Pele/lesões , Cicatrização/efeitos dos fármacos , Animais , Dexametasona/uso terapêutico , Feminino , Camundongos , Oxindóis , Propionatos , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores de Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Receptores do Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Pele/patologia , Resistência à Tração/efeitos dos fármacos
9.
J Pharmacol Exp Ther ; 316(3): 1080-7, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16299187

RESUMO

Idiosyncratic drug toxicity refers to toxic reactions occurring in a small subset of patients and usually cannot be predicted during preclinical or early phases of clinical trials. One hypothesis for the pathogenesis of hepatic idiosyncratic drug reactions is that, in certain individuals, underlying inflammation results in sensitization of the liver, such that injury occurs from an agent that typically would not cause hepatotoxicity at a therapeutic dose. We explored this possibility by cotreating rats with nonhepatotoxic doses of bacterial lipopolysaccharide (LPS) and trovafloxacin (TVX), a drug that caused idiosyncratic hepatotoxicity in humans. The combination of LPS and TVX resulted in hepatotoxicity in rats, as determined by increases in serum alanine aminotransferase activity and hepatocellular necrosis, which were not observed with either agent alone. In contrast, treatment with LPS and levofloxacin, a fluoroquinolone without human idiosyncratic liability, did not result in these changes. Liver gene expression analysis identified unique changes induced by the combination of TVX and LPS, including enhanced expression of chemokines, suggestive of liver neutrophil (PMN) accumulation and activation. Consistent with a role for PMN in the hepatotoxicity induced by LPS/TVX, prior depletion of PMN attenuated the liver injury. The results suggest that gene expression profiles predictive of idiosyncratic liability can be generated in rats cotreated with LPS and drug. Furthermore, they identify gene expression changes that could be explored as biomarkers for idiosyncratic toxicity and lead to enhanced understanding of the mechanism(s) underlying hepatotoxicity induced by TVX.


Assuntos
Quimiocinas/fisiologia , Fluoroquinolonas/toxicidade , Lipopolissacarídeos/farmacologia , Fígado/efeitos dos fármacos , Naftiridinas/toxicidade , Neutrófilos/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos , Alanina Transaminase/sangue , Animais , Quimiocinas CXC/fisiologia , Sinergismo Farmacológico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/fisiologia , Interleucina-6/fisiologia , Fígado/patologia , Masculino , Ratos , Ratos Sprague-Dawley
10.
Toxicol Pathol ; 33(1): 118-26, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15805063

RESUMO

Hepatic enzyme inducers such as phenobarbital are often nongenotoxic rodent hepatocarcinogens. Currently, nongenotoxic hepatocarcinogens can only be definitively identified through costly and extensive long-term, repeat-dose studies (e.g., 2-year rodent carcinogenicity assays). Although liver tumors caused by these compounds are often not found to be relevant to human health, the mechanism(s) by which they cause carcinogenesis are not well understood. Toxicogenomic technologies represent a new approach to understanding the molecular bases of toxicological liabilities such asnongenotoxic carcinogenicity early in the drug discovery/development process. Microarrays have been used to identify mechanistic molecular markers of nongenotoxic rodent hepatocarcinogenesis in short-term, repeat-dose preclinical safety studies. However, the initial "noise" of early adaptive changes may confound mechanistic interpretation of transcription profiling data from short-term studies, and the molecular processes triggered by treatment with a xenobiotic agent are likely to change over the course of long-term treatment. Here, we describe the use of a differential display technology to understand the molecular mechanisms related to 13 weeks of dosing with the prototype rodent nongenotoxic hepatocarcinogen, phenobarbital. These findings implicate a continuing role for oxidative stress in nongenotoxic carcinogenicity.An Excel data file containing raw data is available in full at http://taylorandfrancis.metapress.com/openurl.asp?genre=journal&issn=0192-6233. Click on the issue link for 33(1), then select this article. A download option appears at the bottom of this abstract. The file contains raw data for all gene changes detected by AFLP, including novel genes and genes of unknown function; sequences of detected genes; and animal body and liver weight ratios. In order to access the full article online, you must either have an individual subscription or a member subscription accessed through www.toxpath.org.


Assuntos
Expressão Gênica/efeitos dos fármacos , Fígado/efeitos dos fármacos , Neoplasias/etiologia , Estresse Oxidativo/efeitos dos fármacos , Fenobarbital/toxicidade , Testes de Toxicidade Aguda , Animais , Fígado/metabolismo , Masculino , Modelos Biológicos , Polimorfismo de Fragmento de Restrição , Ratos , Ratos Endogâmicos
11.
Chem Res Toxicol ; 17(4): 463-70, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15089088

RESUMO

Currently, the only way to identify nongenotoxic hepatocarcinogens is through long-term repeat dose studies such as the 2 year rodent carcinogenicity assay. Such assays are both time consuming and expensive and require large amounts of active pharmaceutical or chemical ingredients. Thus, the results of the 2 year assay are not known until very late in the discovery and development process for new pharmaceutical entities. Although in many cases nongenotoxic carcinogenicity in rodents is considered to be irrelevant for humans, a positive finding in a 2 year carcinogenicity assay may increase the number of studies to demonstrate the lack of relevance to humans, delay final submission and subsequent registration of a product, and may result in a "black box" carcinogenicity warning on the label. To develop early identifiers of carcinogenicity, we applied transcription profiling using several prototype rodent genotoxic and nongenotoxic carcinogens, as well as two noncarcinogenic hepatotoxicants, in a 5 day repeat dose in vivo toxicology study. Fluorescent-labeled probes generated from liver mRNA prepared from male Sprague-Dawley rats treated with one of three dose levels of bemitradine, clofibrate, doxylamine, methapyrilene, phenobarbital, tamoxifen, 2-acetylaminofluorene, 4-acetylaminofluorene, or isoniazid were hybridized against rat cDNA microarrays. Correlation of the resulting data with an estimated carcinogenic potential of each compound and dose level identified several candidate molecular markers of rodent nongenotoxic carcinogenicity, including transforming growth factor-beta stimulated clone 22 and NAD(P)H cytochrome P450 oxidoreductase.


Assuntos
Carcinógenos/toxicidade , Transformação Celular Neoplásica , Perfilação da Expressão Gênica , Análise de Sequência com Séries de Oligonucleotídeos , Animais , Bioensaio/métodos , Masculino , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley
12.
Prostate ; 58(4): 366-73, 2004 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-14968437

RESUMO

BACKGROUND: Bone metastases of prostate carcinoma are associated with osteoblastic metastases. Tumor-derived factors, such as parathyroid hormone-related protein (PTHrP), may promote the development of osteoblastic metastases. We examined the effect of transforming growth factor-beta1 (TGF beta 1) on PTHrP mRNA expression and PTHrP secretion in normal canine prostate epithelial cells (PEC) and stromal cells (PSC), and in canine prostate carcinoma cells (PCC). METHODS: Primary cultures of PEC, PSC, and PCC were produced. The effect of TGF beta 1 on PTHrP mRNA expression was measured by Northern blot, and secretion of PTHrP into culture medium was measured by immunoradiometric assay (IRMA). Degradation of recombinant-human PTHrP (rhPTHrP) (1-84) inoculated in prostate cell cultures was measured over 24 hr. Arginine esterase (AE) activity in tissue and conditioned medium was also measured. RESULTS: TGF beta 1 increased PTHrP mRNA expression in a time- and dose-dependent manner in PEC and in PCC. TGF beta 1 decreased PTHrP mRNA in PSC. TGF beta 1 significantly increased PTHrP secretion (P < or = 0.05) into PEC but not PSC conditioned medium. rhPTHrP was significantly (P < or = 0.05) degraded in PEC conditioned medium as compared to PSC conditioned medium. AE activity was present in prostate and prostate carcinoma tissue, but not in conditioned medium from PEC or PSC. CONCLUSIONS: TGF beta 1 increased PTHrP mRNA expression in canine PEC and PCC, and decreased expression in PSC. This regulatory pathway may be important in the pathogenesis of osteoblastic metastases.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Proteína Relacionada ao Hormônio Paratireóideo/genética , Próstata/fisiologia , Neoplasias da Próstata/fisiopatologia , RNA Mensageiro/genética , Células Estromais/citologia , Fator de Crescimento Transformador beta/farmacologia , Animais , Cães , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Células Epiteliais/fisiologia , Masculino , Próstata/citologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Células Estromais/efeitos dos fármacos , Células Estromais/patologia , Células Estromais/fisiologia
13.
J Invest Dermatol ; 121(4): 853-61, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14632205

RESUMO

The cyclooxygenase isoforms, COX-1 and COX-2, are involved in the biosynthesis of prostaglandin E2, a major prostaglandin involved in epidermal homeostasis and repair. Cancer originating in the epidermis can develop when keratinocyte proliferation and apoptosis become dysregulated, resulting in sustained epidermal hyperplasia. COX-2 inhibitors, which demonstrate significant in vivo selectivity relative to COX-1, suppress both ultraviolet-induced epidermal tumor development and progression, suggesting that prostaglandin regulation of keratinocyte biology is involved in the pathogenesis of epidermal neoplasia. In this study, we characterized the expression of COX-1 and COX-2, as well as keratinocyte proliferation, differentiation, and apoptosis, following acute ultraviolet irradiation in the hairless SKH-1 mouse. Following acute ultraviolet exposure, COX-2 expression was predominantly induced in the basal keratinocyte layer coincident with an increase in keratinocyte proliferation and apoptosis. The role of COX-2 was further evaluated using a selective COX-2 inhibitor, SC-791, as well as the traditional nonsteroidal COX inhibitor, indomethacin. Following acute ultraviolet irradiation, inhibition of COX-2 with either inhibitor decreased epidermal keratinocyte proliferation. Likewise, keratinocyte apoptosis was increased with COX-2 inhibition, particularly in the proliferating basal keratinocyte layer. There was also a modest inhibition of keratinocyte differentiation. These data suggest that COX-2 expression is probably necessary for keratinocyte survival and proliferation occurring after acute ultraviolet irradiation. We hypothesize that selective COX-2 inhibition, as described herein, may lead to enhanced removal of ultraviolet-damaged keratinocytes, thereby decreasing malignant transformation in the epidermis.


Assuntos
Células Epidérmicas , Isoenzimas/metabolismo , Queratinócitos/enzimologia , Queratinócitos/efeitos da radiação , Prostaglandina-Endoperóxido Sintases/metabolismo , Neoplasias Cutâneas/prevenção & controle , Doença Aguda , Animais , Divisão Celular/fisiologia , Divisão Celular/efeitos da radiação , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase/farmacologia , Epiderme/enzimologia , Epiderme/efeitos da radiação , Feminino , Isoenzimas/antagonistas & inibidores , Queratinócitos/citologia , Camundongos , Camundongos Pelados , Dermatopatias/tratamento farmacológico , Dermatopatias/metabolismo , Neoplasias Cutâneas/metabolismo , Raios Ultravioleta/efeitos adversos
14.
Toxicol Pathol ; 31(4): 417-31, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12851107

RESUMO

Peroxisome proliferators such as the fibrates act via the peroxisome proliferator activated receptor (PPAR)-alpha as hypolipidemic agents. Many peroxisome proliferators are also nongenotoxic hepatic carcinogens and hepatotoxicants in rodents. We performed transcription profiling using cDNA microarrays on livers of rats treated for 5 days with 3 doses of the peroxisome proliferator clofibrate. All 3 doses had hepatic effects as assessed by liver to body weight ratio, alanine aminotransferase (ALT) increases and histopathology examination. Analysis of the transcription profiling data identified changes in the expression of many genes within several mechanistic pathways that support existing hypotheses regarding peroxisome proliferator mediated carcinogenicity. Additionally, the transcription profiling, histopathology, and clinical chemistry results suggested a biphasic response to clofibrate. These findings provide insight into the pathogenesis of toxic and carcinogenic effects of clofibrate in rodents and demonstrate the ability of cDNA microarrays to provide information regarding mechanisms of toxicity identified during the drug development process.


Assuntos
Clofibrato/toxicidade , Perfilação da Expressão Gênica , Fígado/efeitos dos fármacos , Proliferadores de Peroxissomos/toxicidade , Alanina Transaminase/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Fígado/patologia , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Análise de Componente Principal , Ratos
15.
Thyroid ; 13(2): 133-40, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12699587

RESUMO

The sodium iodide symporter (NIS) mediates iodide uptake in thyroid cells and enables the effective radioiodide treatment of thyroid cancers. There is much interest in facilitating radioiodide therapy in other cancers by NIS gene transfer. This study showed that exogenous NIS expression decreased MATLyLu rat prostatic adenocarcinoma cell growth. Tumor growth and metastatic progression were significantly delayed in syngeneic rats injected with mixed or clonal populations of MATLyLu-NIS cells compared to rats with control tumors. MATLyLu-NIS tumors in nude mice had a lower, albeit not statistically significant, growth rate than control tumors. The Ki-67 labeling index in NIS-positive areas was lower than in NIS-negative areas of rat tumors derived from a mixed population of MATLyLu-NIS cells. Growth of clonal populations of MATLyLu-NIS cells was delayed in vitro. These results demonstrate that NIS expression inhibits MATLyLu cell growth, thereby providing an additional potential benefit of NIS-mediated gene therapy for cancer.


Assuntos
Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Simportadores/biossíntese , Adenocarcinoma/ultraestrutura , Animais , Divisão Celular/efeitos dos fármacos , Células Clonais , DNA/genética , Humanos , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Nus , Microscopia Eletrônica , Metástase Neoplásica/tratamento farmacológico , Metástase Neoplásica/patologia , Transplante de Neoplasias , Neoplasias da Próstata/ultraestrutura , Ratos , Retroviridae/genética , Transplante Heterólogo , Células Tumorais Cultivadas
16.
Kidney Int ; 63(5): 1791-800, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12675855

RESUMO

BACKGROUND: We evaluated the role of aldosterone as a mediator of renal inflammation and fibrosis in a rat model of aldosterone/salt hypertension using the selective aldosterone blocker, eplerenone. METHODS: Unnephrectomized, Sprague-Dawley rats were given 1% NaCl (salt) to drink and randomized to receive treatment for 28 days: vehicle infusion (control); 0.75 microg/hour aldosterone subcutaneous infusion; or aldosterone infusion + 100 mg/kg/day oral dose of eplerenone. Blood pressure and urinary albumin were measured and kidneys were evaluated histologically. Renal injury, inflammation, and fibrosis were assessed by immunohistochemistry, in situ hybridization, and reverse transcription-polymerase chain reaction (RT-PCR). RESULTS: Aldosterone/salt induced severe hypertension compared to controls (220 +/- 4 mm Hg vs. 131 +/- 4 mm Hg, P < 0.05), which was partially attenuated by eplerenone (179 +/- 4 mm Hg, P < 0.05). In aldosterone/salt treated rats, renal histopathologic evaluation revealed severe vascular and glomerular sclerosis, fibrinoid necrosis and thrombosis, interstitial leukocyte infiltration, and tubular damage and regeneration. Aldosterone/salt increased circulating osteopontin (925.0 +/- 80.2 ng/mL vs. 53.6 +/- 6.3 ng/mL) and albuminuria (75.8 +/- 10.9 mg/24 hours vs. 13.2 +/- 3.0 mg/24 hours) compared to controls and increased expression of proinflammatory molecules. Treatment with eplerenone reduced systemic osteopontin (58.3 +/- 4.2 ng/mL), albuminuria (41.5 +/- 7.2 mg/24 hours), and proinflammatory gene expression: osteopontin (OPN), monocyte chemoattractant protein-1 (MCP-1), interleukin-6 (IL-6), and interleukin-1beta (IL-1beta). CONCLUSION: These findings indicate that aldosterone/salt-induced renal injury and fibrosis has inflammatory components involving macrophage infiltration and cytokine up-regulation. Attenuation of renal damage and inflammation by eplerenone supports the protective effects of aldosterone blockade in hypertensive renal disease.


Assuntos
Aldosterona/farmacologia , Hipertensão Renal/imunologia , Nefrite/induzido quimicamente , Nefrite/imunologia , Cloreto de Sódio/farmacologia , Espironolactona/análogos & derivados , Animais , Pressão Sanguínea , Citocinas/metabolismo , Eplerenona , Fibrose , Hipertensão Renal/tratamento farmacológico , Hipertensão Renal/patologia , Imuno-Histoquímica , Hibridização In Situ , Rim/imunologia , Rim/patologia , Macrófagos/patologia , Masculino , Nefrite/patologia , Ratos , Ratos Sprague-Dawley , Espironolactona/farmacologia
17.
J Pathol ; 199(2): 221-8, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12533835

RESUMO

The terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick-end labelling (TUNEL) technique has been extensively used for the detection and quantification of apoptosis in histological tissue sections. However, the interpretation and specificity of this assay have been controversial. With accumulating knowledge of the molecular mechanisms of cell death and the discovery of the caspases as key mediators of apoptosis, more direct and earlier measurements of apoptosis in tissue sections have emerged. This study, using antibodies that specifically recognize activated caspase-3 and caspase-cleaved cytokeratin (CK) 18, evaluated whether immunohistochemical stains would improve the detection and quantification of apoptosis in tissue sections, compared with the TUNEL assay. Tumour xenografts of the prostate cancer cell line PC-3 were used as an example, since these tissues contain large numbers of cells undergoing apoptosis. Apoptotic cells were quantified and apoptotic indices were calculated by computer-assisted image analysis following identification of apoptotic cells by morphological analysis, the TUNEL assay, activated caspase-3 and cleaved CK18 immunohistochemistry. The results indicated that activated caspase-3 immunohistochemistry was an easy, sensitive, and reliable method for detecting and quantifying apoptosis in this model. An excellent correlation (R = 0.89) between the apoptotic indices obtained using activated caspase-3 and cleaved CK18 immunostaining was observed. A good correlation (R = 0.75) between the apoptotic indices obtained using activated caspase-3 immunostaining and the TUNEL assay was also found. Activated caspase-3 immunohistochemistry is therefore recommended for the detection and quantification of apoptosis in tissue sections.


Assuntos
Apoptose , Caspases/análise , Marcação In Situ das Extremidades Cortadas/métodos , Queratinas/análise , Animais , Caspase 3 , Contagem de Células , Feminino , Processamento de Imagem Assistida por Computador/métodos , Imuno-Histoquímica/métodos , Camundongos , Camundongos Nus , Sensibilidade e Especificidade , Transplante Heterólogo , Células Tumorais Cultivadas/patologia
18.
Comp Med ; 52(6): 568-71, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12540173

RESUMO

Epididymal cribriform hyperplasia (ECH) is a variant of normal epididymal histologic features in men, and has also been reported in rats, mice, dogs, cats, and bulls. The epididymal change has been associated with aging, testicular atrophy, cryptorchidism, and germ cell tumors. Epididymal cribriform hyperplasia was observed in p53 homozygous knockout mice on a mixed 129/Sv-FVB/N background, but not in wild-type or heterozygous mice. The aim of the study reported here was to determine the prevalence and characterize the morphologic, immunohistochemical, and ultrastructural features of ECH in these mice. Epididymal cribriform hyperplasia was present in 88% (72/82) of male mice ranging in age from seven to 65 weeks. The lesion was characterized microscopically by epithelial cells with atypical hyperchromatic nuclei, vacuolization, intratubular lumina formation, infrequent apoptosis, and rare mitotic figures. In contrast to germ cells, the cells of ECH did not express alpha-fetoprotein, carcinoembryonic antigen, or S-100. Ultrastructurally, the cells were pleomorphic with stereocilia at their apical borders and within intratubular lumina, and were supported by a basement membrane. Although 14% (10/72) of mice had concomitant testicular neoplasia, ECH did not appear to be a preneoplastic change. Investigators using these mice for modeling human disease should be aware of the background prevalence of this lesion.


Assuntos
Núcleo Celular/patologia , Epididimo/patologia , Doenças Testiculares/patologia , Proteína Supressora de Tumor p53/genética , Fatores Etários , Animais , Cruzamento , DNA/análise , Primers do DNA/química , Modelos Animais de Doenças , Epididimo/metabolismo , Células Epiteliais/ultraestrutura , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Genótipo , Homozigoto , Hiperplasia/patologia , Técnicas Imunoenzimáticas , Masculino , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase , Doenças Testiculares/metabolismo , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA