Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Pharmacol Exp Ther ; 384(1): 35-51, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35809898

RESUMEN

MicroRNAs (miRNAs) are involved in the development of human malignancies, and cells have the ability to secrete these molecules into extracellular compartments. Thus, cell-free miRNAs (circulating miRNAs) can potentially be used as biomarkers to evaluate pathophysiological changes. Although circulating miRNAs have been proposed as potential noninvasive tumor biomarkers for diagnosis, prognosis, and response to therapy, their routine application in the clinic is far from being achieved. This review focuses on the recent progress regarding the value of circulating miRNAs as noninvasive biomarkers, with specific consideration of their relevant clinical applications. In addition, we provide an in-depth analysis of the technical challenges that impact the assessment of circulating miRNAs. We also highlight the significance of integrating circulating miRNAs with the standard laboratory biomarkers to boost sensitivity and specificity. The current status of circulating miRNAs in clinical trials as tumor biomarkers is also covered. These insights and general guidelines will assist researchers in experimental practice to ensure quality standards and repeatability, thus improving future studies on circulating miRNAs. SIGNIFICANCE STATEMENT: Our review will boost the knowledge behind the inconsistencies and contradictory results observed among studies investigating circulating miRNAs. It will also provide a solid platform for better-planned strategies and standardized techniques to optimize the assessment of circulating cell-free miRNAs.


Asunto(s)
Biomarcadores de Tumor , MicroARN Circulante , Neoplasias , Humanos , Biomarcadores de Tumor/sangre , MicroARN Circulante/sangre , Neoplasias/sangre , Neoplasias/diagnóstico , Pronóstico
2.
Mar Drugs ; 21(4)2023 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-37103355

RESUMEN

Metastatic castration-resistant prostate cancer (mCRPC) cells can de novo biosynthesize their own cholesterol and overexpress proprotein convertase subtilisin/kexin type 9 (PCSK9). PCSK9 proved to contribute to mCRPC cell motility since PCSK9 knockdown (KD) in mCRPC CWR-R1ca cells led to notable reductions in cell migration and colony formation. Human tissue microarray results proved a higher immunohistoscore in patients ≥ 65 years old, and PCSK9 proved to be expressed higher at an early Gleason score of ≤7. The fermentation product pseurotin A (PS) suppressed PCSK9 expression, protein-protein interactions with LDLR, and breast and prostate cancer recurrences. PS suppressed migration and colony formation of the CWR-R1ca cells. The progression and metastasis of the CWR-R1ca-Luc cells subcutaneously (sc) xenografted into male nude mice fed a high-fat diet (HFD, 11% fat content) showed nearly 2-fold tumor volume, metastasis, serum cholesterol, low-density lipoprotein cholesterol (LDL-C), prostate-specific antigen (PSA), and PCSK9 levels versus mice fed a regular chow diet. Daily oral PS 10 mg/kg treatments prevented the locoregional and distant tumor recurrence of CWR-R1ca-Luc engrafted into nude mice after primary tumor surgical excision. PS-treated mice showed a significant reduction in serum cholesterol, LDL-C, PCSK9, and PSA levels. These results comprehensively validate PS as an mCRPC recurrence-suppressive lead by modulating the PCSK9-LDLR axis.


Asunto(s)
Proproteína Convertasa 9 , Neoplasias de la Próstata Resistentes a la Castración , Humanos , Masculino , Ratones , Animales , Anciano , Proproteína Convertasa 9/genética , Proproteína Convertasa 9/metabolismo , Ratones Desnudos , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Serina Endopeptidasas/metabolismo , Proproteína Convertasas/metabolismo , Antígeno Prostático Específico , Receptores de LDL/genética , Receptores de LDL/metabolismo , Colesterol
3.
Environ Toxicol ; 38(2): 266-277, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36447373

RESUMEN

Prodigiosin (PDG) is a bacterial metabolite with numerous biological and pharmaceutical properties. Exposure to aluminium is considered a root etiological factor in the pathological progress of Alzheimer's disease (AD). Here, in this investigation, we explored the neuroprotective potential of PDG against aluminium chloride (AlCl3 )-mediated AD-like neurological alterations in rats. For this purpose, rats were gavaged either AlCl3 (100 mg/kg), PDG (300 mg/kg), or both for 42 days. As a result of the analyzes performed on the hippocampal tissue, it was observed that AlCl3 induced biochemical, molecular, and histopathological changes like those related to AD. PDG pre-treatment significantly decreased acetylcholinesterase activity and restored the levels of brain-derived neurotrophic factor, monoamines (dopamine, norepinephrine, and serotonin), and transmembrane protein (Na+ /K+ -ATPase). Furthermore, PDG boosted the hippocampal antioxidant capacity, as shown by the increased superoxide dismutase, catalase, glutathione peroxidase, glutathione reductase, and glutathione contents. These findings were accompanied by decreases in malondialdehyde and nitric oxide levels. The antioxidant effect may promote the upregulation of the expression of antioxidant genes (Nrf2 and HO-1). Moreover, PDG exerted notable anti-inflammatory effects via the lessening of interleukin-1 beta, tumor necrosis factor-alpha, cyclooxygenase-2, nuclear factor kappa B, and decreases in the gene expression of inducible nitric oxide synthase. In addition, noteworthy decreases in pro-apoptotic (Bax and caspase-3) levels and increases in anti-apoptotic (Bcl-2) biomarkers suggested an anti-apoptotic effect of PDG. In support, the hippocampal histological examination validated the aforementioned changes. To summarize, the promising neuromodulatory, antioxidative, anti-inflammatory, and anti-apoptotic activities of PDG establish it as a potent therapeutic option for AD.


Asunto(s)
Enfermedad de Alzheimer , Fármacos Neuroprotectores , Animales , Ratas , Acetilcolinesterasa/metabolismo , Cloruro de Aluminio/toxicidad , Cloruro de Aluminio/uso terapéutico , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Antiinflamatorios/farmacología , Antioxidantes/metabolismo , Glutatión/metabolismo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Estrés Oxidativo , Prodigiosina/metabolismo , Prodigiosina/farmacología , Prodigiosina/uso terapéutico
4.
Int J Mol Sci ; 24(9)2023 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-37176114

RESUMEN

The adaptive acquisition of resistance to BRAF and MEK inhibitor-based therapy is a common feature of melanoma cells and contributes to poor patient treatment outcomes. Leveraging insights from a proteomic study and publicly available transcriptomic data, we evaluated the predictive capacity of a gene panel corresponding to proteins differentially abundant between treatment-sensitive and treatment-resistant cell lines, deciphering predictors of treatment resistance and potential resistance mechanisms to BRAF/MEK inhibitor therapy in patient biopsy samples. From our analysis, a 13-gene signature panel, in both test and validation datasets, could identify treatment-resistant or progressed melanoma cases with an accuracy and sensitivity of over 70%. The dysregulation of HMOX1, ICAM, MMP2, and SPARC defined a BRAF/MEK treatment-resistant landscape, with resistant cases showing a >2-fold risk of expression of these genes. Furthermore, we utilized a combination of functional enrichment- and gene expression-derived scores to model and identify pathways, such as HMOX1-mediated mitochondrial stress response, as potential key drivers of the emergence of a BRAF/MEK inhibitor-resistant state in melanoma cells. Overall, our results highlight the utility of these genes in predicting treatment outcomes and the underlying mechanisms that can be targeted to reduce the development of resistance to BRAF/MEK targeted therapy.


Asunto(s)
Melanoma , Proteínas Proto-Oncogénicas B-raf , Humanos , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteómica , Resistencia a Antineoplásicos/genética , Línea Celular Tumoral , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo
5.
J Cell Mol Med ; 26(22): 5624-5633, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36300880

RESUMEN

Curcumin is a known epigenetic modifier that demonstrated antitumor effect in different types of cancer. The poor solubility and metabolic stability are major drawbacks that limit its development as an antitumor agent. Dimethoxycurcumin (DMC) is a more soluble and stable curcumin analog. In this study, we compared the effect of both drugs on a variety of histone posttranslational modifications and on the activity of histone lysine methyltransferase (HKMTs) and demethylase (HKDMTs) enzymes that target the H3K4, H3K9 and H3K27 epigenetic marks. Mass spectrometry was used to quantitate the changes in 95 histone posttranslational modifications induced by curcumin or DMC. The effect of both drugs on the enzymatic activity of HKMTs and HKDMs was measured using an antibody-based assay. Mass spectrometry analysis showed that curcumin and DMC modulated several histone modifications. Histone changes were not limited to lysine methylation and acetylation but included arginine and glutamine methylation. Only few histone modifications were similarly changed by both drugs. On the contrary, the effect of both drugs on the activity of HKMTs and HKDMs was very similar. Curcumin and DMC inhibited the HKMTs enzymes that target the H3K4, H3K9 and H3K27 marks and increased the activity of the HKDMs enzymes LSD1, JARID and JMJD2. In conclusion, we identified novel enzymatic targets for both curcumin and DMC that support their use and development as epigenetic modifiers in cancer treatment. The multiple targets modulated by both drugs could provide a therapeutic advantage by overcoming drug resistance development.


Asunto(s)
Curcumina , Leucemia , Humanos , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Curcumina/farmacología , Leucemia/tratamiento farmacológico
6.
Cancer Control ; 29: 10732748221074051, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35067084

RESUMEN

INTRODUCTION: The prototype DNA hypomethylating agents 5-azacytidine (5AC) and decitabine (DAC) are currently FDA-approved for treatment of blood and bone marrow disorders like myelodysplastic syndrome. 5AC and DAC are considered similar drugs and were shown to induce histone modifications that modulate gene expression. The aim of this study is to compare the effect of both drugs on histone acetylation and methylation at multiple histone amino acids residues. METHODS: Mass spectrometry was used to compare the effect of both drugs on 95 different histone posttranslational modifications (PTMs) in leukemia cells. ChIP-Seq analysis was used to compare the impact of both drugs on the genome-wide acetylation of the H3K9 mark using primary leukemia cells from six de-identified AML patients. RESULTS: Both DAC and 5AC induced histone PTMs in different histone isoforms like H1.4, H2A, H3, H3.1, and H4. Changes in both histone methylation and acetylation were observed with both drugs; however, there were distinct differences in the histone modifications induced by the two drugs. Since both drugs were shown to increase the activity of the HDAC SIRT6 previously, we tested the effect of 5AC on the acetylation of H3K9, the physiological substrate SIRT6, using ChIP-Seq analysis and compared it to the previously published DAC-induced changes. Significant H3K9 acetylation changes (P< .05) were detected at 925 genes after 5AC treatment vs only 182 genes after DAC treatment. Nevertheless, the gene set modified by 5AC was different from that modified by DAC with only ten similar genes modulated by both drugs. CONCLUSION: Despite similarity in chemical structure and DNA hypomethylating activity, 5AC and DAC induced widely different histone PTMs and considering them interchangeable should be carefully evaluated. The mechanism of these histone PTM changes is not clear and may involve modulation of the activity or the expression of the enzymes inducing histone PTMs.


Asunto(s)
Acetilación/efectos de los fármacos , Azacitidina/farmacología , Metilación de ADN/efectos de los fármacos , Decitabina/farmacología , Histonas/efectos de los fármacos , Línea Celular Tumoral , Humanos , Leucemia/tratamiento farmacológico , Procesamiento Proteico-Postraduccional/efectos de los fármacos
7.
Arch Pharm (Weinheim) ; 355(6): e2100327, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35285986

RESUMEN

Two new series of coumarin and benzofuran derivatives were designed, synthesized, and assessed for their in vitro and in vivo antitumor activities against breast cancer. Compounds 8, 9, 14, 15, and 17 exhibited the best antiproliferative activities (IC50 : 0.07-2.94 µM) against the MCF-7 cell line, compared with lapatinib (IC50 : 4.69 µM). Compound 14, with the most potent cytotoxic activity against MCF-7 cells, was capable of enhancing preG1 apoptosis and triggering cell cycle arrest at the G2/M phase. The kinase inhibitory activity of compound 14 against a panel of 22 kinases was examined to reveal multikinase inhibition within -39% to -97%. Furthermore, compound 14 exhibited potent in vivo Ehrlich (mammary adenocarcinoma) tumor regression, positive caspase-3, and negative EGFR immunoreaction, and was capable of elevating the catalase level. The physicochemical properties and pharmacokinetic parameters of compound 14 were investigated in silico for its druglikeness.


Asunto(s)
Antineoplásicos , Benzofuranos , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis , Benzofuranos/química , Benzofuranos/farmacología , Línea Celular Tumoral , Proliferación Celular , Cumarinas/farmacología , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Estructura Molecular , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Relación Estructura-Actividad
8.
Bioorg Med Chem ; 27(7): 1308-1319, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30792101

RESUMEN

Two new series of furochromone and benzofuran derivatives were designed, synthesized and evaluated for their in vitro anticancer activity against MCF-7 and MDA231 breast cancer cell lines. Compounds 5, 6, 7, 9, 15a, 16, 17a and 18 exhibited the best antiproliferative activities with IC50 values ranging from 1.19 to 2.78 µM against MCF-7 superior to lapatinib as reference standard (IC50; 4.69 µM). Compounds 15a and 18 revealed significant cytotoxic activity against MCF-7 and MDA231, therefore their inhibitory potencies against p38α MAP kinase were evaluated. Remarkably they exhibited significant IC50 of 0.04 µM comparable to SB203580 (IC50; 0.50 µM) as a reference standard. These promising results of cytotoxic activity and significant inhibition of p38α MAP kinase, were confirmed by exploring the effect of benzofuran derivative (18) on the apoptotic induction and cell cycle progression of MCF-7 cell line. Compound 18 induced preG1 apoptosis and cell growth arrest at G2/M phase preventing the mitotic cycle. Moreover it activated the caspase-7 which executes apoptosis. Molecular docking study was carried out using GOLD program to predict the mode of binding interaction of the synthesized compounds into the target p38α MAPK. Additionally, the physicochemical properties and ADME parameters of compound 18 were examined in silico to investigate its drug-likeness.


Asunto(s)
Antineoplásicos/farmacología , Productos Biológicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Diseño de Fármacos , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Productos Biológicos/síntesis química , Productos Biológicos/química , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Compuestos Heterocíclicos/química , Compuestos Heterocíclicos/farmacología , Humanos , Células MCF-7 , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Modelos Moleculares , Estructura Molecular , Oxígeno/química , Oxígeno/farmacología , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad , Células Tumorales Cultivadas
9.
Biomarkers ; 22(7): 621-628, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27788588

RESUMEN

CONTEXT: The incidence rate of hepatocellular carcinoma (HCC) is higher in developing countries, and most cases are associated with chronic hepatitis C virus (HCV) infection. OBJECTIVE: To evaluate the circulating proteins as liver biomarkers for the identification of HCC associated with HCV infection in Egyptian patients using LC-MS/MS analysis. METHODS: Blood sera were collected from 31 HCC patients and the fractionated proteins were subjected to LC-MS/MS analysis. Protein candidates were validated by enzyme-linked immunosorbent assay (ELISA). RESULTS: Thirty-three proteins were significantly identified in the sera of HCC patients with persistent HCV infection. These proteins are involved in several biological processes including acute phase response, complement activation, hemostasis process and lipid metabolism. The level of lectin galactoside-binding soluble 3 binding protein (LGALS3BP), Kininogen-1 (KNG1), serum amyloid A2 (SAA2) and paraoxonase 1 (PON1) and alpha-fetoprtoein (AFP) were elevated in serum. CONCLUSION: In HCC patients with chronic HCV infection, we identified a group of differentially expressed circulating proteins involved in regulating different cellular mechanisms.


Asunto(s)
Biomarcadores de Tumor/sangre , Carcinoma Hepatocelular/sangre , Hepatitis C Crónica/sangre , Neoplasias Hepáticas/sangre , Carcinoma Hepatocelular/complicaciones , Egipto , Hepatitis C Crónica/complicaciones , Humanos , Proteínas/análisis , Proteómica/métodos
10.
J Cell Mol Med ; 20(2): 266-86, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26578344

RESUMEN

Melanoma is characterized by dysregulated intracellular signalling pathways including an impairment of the cell death machinery, ultimately resulting in melanoma resistance, survival and progression. This explains the tumour's extraordinary resistance to the standard treatment. Imiquimod is a topical immune response modifier (imidazoquinoline) with both antiviral and antitumour activities. The mechanism by which imiquimod triggers the apoptosis of melanoma cells has now been carefully elucidated. Imiquimod-induced apoptosis is associated with the activation of apoptosis signalling regulating kinase1/c-Jun-N-terminal kinase/p38 pathways and the induction of endoplasmic stress characterized by the activation of the protein kinase RNA-like endoplasmic reticulum kinase signalling pathway, increase in intracellular Ca(2+) release, degradation of calpain and subsequent cleavage of caspase-4. Moreover, imiquimod triggers the activation of NF-κB and the expression of the inhibitor of apoptosis proteins (IAPs) such as, X-linked IAP (XIAP) together with the accumulation of reactive oxygen species (ROS). Also, imiquimod triggers mitochondrial dysregulation characterized by the loss of mitochondrial membrane potential (Δψm), the increase in cytochrome c release, and cleavage of caspase-9, caspase-3 and poly(ADP-ribose) polymerase (PARP). Inhibitors of specific pathways, permit the elucidation of possible mechanisms of imiquimod-induced apoptosis. They demonstrate that inhibition of NF-kB by the inhibitor of nuclear factor kappa-B kinase (IKK) inhibitor Bay 11-782 or knockdown of XIAP induces melanoma apoptosis in cells exposed to imiquimod. These findings support the use of either IKK inhibitors or IAP antagonists as adjuvant therapies to improve the effectiveness topical imiquimod in the treatment of melanoma.


Asunto(s)
Aminoquinolinas/farmacología , Apoptosis/efectos de los fármacos , Estrés del Retículo Endoplásmico/fisiología , Melanoma/tratamiento farmacológico , FN-kappa B/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Calcio/metabolismo , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Línea Celular Tumoral , Citocromos c/metabolismo , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Humanos , Imiquimod , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Melanoma/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo
11.
Ann Surg Oncol ; 23(9): 2883-8, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27146414

RESUMEN

BACKGROUND: Metadherin (MTDH) is widely recognized as a promising molecular marker for tumor recurrence and poor survival in many cancers. By multiple pathways, MTDH promotes oncogenesis, metastasis, and chemoresistance. This study investigated the role of MTDH in papillary thyroid carcinoma (PTC) to determine its potential association with aggressive clinical and pathologic features, including its relation in tumors harboring a BRAF (V600E) mutation. METHODS: Expression of MTDH was assessed by immunohistochemistry in 96 cases of PTC, including primary thyroid malignancies and lymph node metastases. The status of BRAF (V600E) mutation was determined by real-time polymerase chain reaction. RESULTS: Overexpression of MTDH was observed in 26 % (23/88) of primary PTC cases. High-intensity staining was observed in 75 % (6/8) of lymph nodes with metastatic PTC and moderate staining in 25 % (2/8) of cases. Normal adjacent thyroid tissue and benign thyroid controls were found to have significantly lower MTDH expression than cancer tissue (p < 0.05). Apical staining of MTDH was observed in 19 % of thyroid tumors and not observed in normal thyroid tissue. Interestingly, MTDH expression was associated with extrathyroidal extension (p < 0.05) and not associated with age, gender, overall tumor stage, or BRAF (V600E) mutation status. CONCLUSION: In a subset of PTC patients, MTDH was overexpressed and associated with extrathyroidal extension. Further studies are warranted to explore the utility of MTDH to improve risk stratification of current molecular panels for PTC.


Asunto(s)
Carcinoma Papilar/metabolismo , Carcinoma Papilar/secundario , Moléculas de Adhesión Celular/metabolismo , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología , Adulto , Carcinoma Papilar/genética , Femenino , Humanos , Metástasis Linfática , Masculino , Proteínas de la Membrana , Persona de Mediana Edad , Invasividad Neoplásica , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas de Unión al ARN , Glándula Tiroides/metabolismo , Neoplasias de la Tiroides/genética
12.
J Surg Res ; 203(2): 407-15, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27363650

RESUMEN

BACKGROUND: The BRAF-V600E mutation is associated with tumor aggressiveness and poor prognosis in melanoma patients. Identification of this mutation is clinically important as we now have Food and Drug Administration-approved targeted therapies, such as BRAF and MEK inhibitors, which have been shown to retard disease progression in these patients. Detection of BRAF-V600E by genetic analysis using polymerase chain reaction is the gold standard method for melanoma cases. However, immunohistochemistry (IHC) using a VE1 antibody is rapidly emerging as a trustworthy method for the determination of mutation status in patients' specimens. Our objective in this study was to assess the reliability of IHC compared with genetic methods for successful identification of BRAF-V600E mutation in melanoma tissue specimens. METHODS: A literature search of PubMed, Web of Science, and Embase was performed for studies comparing IHC with genetic analysis for the detection of BRAF in melanoma patients published through May 28, 2015. Pooled sensitivity, specificity, diagnostic odds ratio, positive, and negative likelihood ratios were calculated using a bivariate model. Logit estimates of sensitivity and specificity with their respective variances were used to plot a hierarchical receiver operating characteristic curve and area under the curve. Heterogeneity was assessed using the Q- and I-squared statistics. RESULTS: An initial literature search resulted in 287 articles. After two independent reviews and consensus-based discussion to resolve disparities, 21 studies involving a total of 1687 cases met the eligibility criteria and were included in the analysis. The pooled sensitivity of IHC for BRAF-V600E detection was 0.96; 95% confidence interval (CI, 0.94-0.98), specificity 1.00; 95% CI (0.97-1.00), positive likelihood ratio 194.2; 95% CI (37.6-1003.3), negative likelihood ratio 0.04; 95% CI (0.02-0.07), and diagnostic odds ratio 5503 (1199-25,263), as compared with genetic analysis. A high heterogeneity was observed between these studies (Q value of 40.17 & I(2) = 95%; 95% CI (91-99, P < 0.001) which may be explained by studies using different cutoff values for labeling IHC as positive. High accuracy of IHC was depicted by area under the curve in the receiver operating characteristic curve which was 0.99; 95 % CI (0.98-1.00). CONCLUSIONS: Meta-analysis demonstrates that IHC is highly sensitive and specific for the detection of BRAF-V600E in melanoma cases. IHC is likely to be useful in BRAF mutation detection because it is highly comparable with the genetic methods. Any negative or low staining cases may be selected to undergo genetic analysis based on other clinical and histopathologic features.


Asunto(s)
Biomarcadores de Tumor/genética , Inmunohistoquímica , Melanoma/genética , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias Cutáneas/genética , Análisis Mutacional de ADN/métodos , Humanos , Modelos Estadísticos , Reacción en Cadena de la Polimerasa , Sensibilidad y Especificidad
13.
J Transl Med ; 13: 233, 2015 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-26183824

RESUMEN

BACKGROUND: We and others have extensively investigated the role of PARP-1 in cell growth and demise in response to pathophysiological cues. Most of the clinical trials on PARP inhibitors are targeting primarily estrogen receptor (ER) negative cancers with BRCA-deficiency. It is surprising that the role of the enzyme has yet to be investigated in ER-mediated cell growth. It is noteworthy that ER is expressed in the majority of breast cancers. We recently showed that the scaffolding protein PDZK1 is critical for 17ß-estradiol (E2)-induced growth of breast cancer cells. We demonstrated that E2-induced PDZK1 expression is indirectly regulated by ER and requires IGF-1 receptor (IGF-1R). METHODS: The breast cancer cell lines MCF-7 and BT474 were used as ER(+) cell culture models. Thieno[2,3-c]isoquinolin-5-one (TIQ-A) and olaparib (AZD2281) were used as potent inhibitors of PARP. PARP-1 knockdown by shRNA was used to show specificity of the effects to PARP-1. RESULTS: In this study, we aimed to determine the effect of PARP inhibition on estrogen-induced growth of breast cancer cells and examine whether the potential effect is linked to PDZK1 and IGF-1R expression. Our results show that PARP inhibition pharmacologically by TIQ-A or olaparib or by PARP-1 knockdown blocked E2-dependent growth of MCF-7 cells. Such inhibitory effect was also observed in olaparib-treated BT474 cells. The effect of PARP inhibition on cell growth coincided with an efficient reduction in E2-induced PDZK1 expression. This effect was accompanied by a similar decrease in the cell cycle protein cyclin D1. PARP appeared to regulate E2-induced PDZK1 at the mRNA level. Such regulation may be linked to a modulation of IGF-1R as PARP inhibition pharmacologically or by PARP-1 knockdown efficiently reduced E2-induced expression of the receptor at the protein and mRNA levels. CONCLUSIONS: Overall, our results show for the first time that PARP regulates E2-mediated cell growth by controlling the ER/IGF-1R/PDZK1 axis. These findings suggest that the relationship between ER, PDZK1, and IGF-1R may be perturbed by blocking PARP function and that PARP inhibitors may be considered in clinical trials on ER(+) cancers.


Asunto(s)
Proteínas Portadoras/metabolismo , Estradiol/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Receptor IGF Tipo 1/metabolismo , Receptores de Estrógenos/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Portadoras/genética , Proliferación Celular/efectos de los fármacos , Ciclina D1 , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Células MCF-7 , Proteínas de la Membrana , Ftalazinas/farmacología , Piperazinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo
14.
J Urol ; 194(3): 820-7, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25676431

RESUMEN

PURPOSE: We sought to develop a reproducible TGF-ß1 injection technique to induce urethral fibrosis in the rat urethra. MATERIALS AND METHODS: A total of 32 male Sprague Dawley® rats weighing 300 to 350 gm were anesthetized with ketamine/xylazine intraperitoneally. Using a 5 mm penoscrotal incision the rat urethra was exposed. In the experimental group varying doses of TGF-ß1 (5, 10 and 25 µg) were injected in each side of the urethral wall. Normal saline infiltration was used in the sham treated group. Rats were sacrificed 2 and 4 weeks following TGF-ß1 injection. Urethral specimens were stained with hematoxylin and eosin, and Masson trichrome, and Western blot evaluations were performed. Normal and strictured urethral tissues from patients were collected and evaluated in the same fashion. RESULTS: There was no evidence of urethral wall thickening or fibrosis in the sham treated group. Varied histological evidence of fibrosis was noted in all experimental groups. There was a significant increase in collagen type I expression 2 weeks after injection of 5, 10 and 25 µg TGF-ß1. Collagen type III expression was significantly increased 2 weeks after injecting 10 and 25 µg of TGF-ß1, which persisted to 28 days after injection. CONCLUSIONS: TGF-ß1 injection can successfully generate a reproducible rat model of urethral spongiofibrosis. This technique is simple, inexpensive and reproducible. Our series is a proof of concept study. Additional studies in larger animals are needed to further confirm our findings.


Asunto(s)
Modelos Animales de Enfermedad , Factor de Crecimiento Transformador beta1/administración & dosificación , Uretra/patología , Estrechez Uretral/inducido químicamente , Animales , Fibrosis/inducido químicamente , Inyecciones , Masculino , Ratas , Ratas Sprague-Dawley
15.
Stem Cells ; 32(4): 983-97, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24715691

RESUMEN

Emerging evidence suggests that mesenchymal stem cells (MSCs) are often recruited to tumor sites but their functional significance in tumor growth and disease progression remains elusive. Herein we report that prostate cancer (PC) cell microenvironment subverts PC patient adipose-derived stem cells (pASCs) to undergo neoplastic transformation. Unlike normal ASCs, the pASCs primed with PC cell conditioned media (CM) formed prostate-like neoplastic lesions in vivo and reproduced aggressive tumors in secondary recipients. The pASC tumors acquired cytogenetic aberrations and mesenchymal-to-epithelial transition and expressed epithelial, neoplastic, and vasculogenic markers reminiscent of molecular features of PC tumor xenografts. Our mechanistic studies revealed that PC cell-derived exosomes are sufficient to recapitulate formation of prostate tumorigenic mimicry generated by CM-primed pASCs in vivo. In addition to downregulation of the large tumor suppressor homolog2 and the programmed cell death protein 4, a neoplastic transformation inhibitor, the tumorigenic reprogramming of pASCs was associated with trafficking by PC cell-derived exosomes of oncogenic factors, including H-ras and K-ras transcripts, oncomiRNAs miR-125b, miR-130b, and miR-155 as well as the Ras superfamily of GTPases Rab1a, Rab1b, and Rab11a. Our findings implicate a new role for PC cell-derived exosomes in clonal expansion of tumors through neoplastic reprogramming of tumor tropic ASCs in cancer patients.


Asunto(s)
Tejido Adiposo/metabolismo , Comunicación Celular , Transformación Celular Neoplásica/metabolismo , Transición Epitelial-Mesenquimal , Exosomas/metabolismo , Neoplasias de la Próstata/metabolismo , Células Madre/metabolismo , Tejido Adiposo/patología , Transformación Celular Neoplásica/patología , Exosomas/patología , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/metabolismo , Neoplasias de la Próstata/patología , ARN Neoplásico/metabolismo , Células Madre/patología
16.
Pancreatology ; 15(1): 34-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25464937

RESUMEN

BACKGROUND: Despite the incidence rate of pancreatic cancer (PC) is uncommon in developing countries, it is considered as one of the most lethal disease. Improving patients' survival requires diagnosis of the disease at early stage. Therefore, it is imperative to identify more specific and sensitive marker(s) to be used for early detection of PC. OBJECTIVES: Our aim is to evaluate the potential role of circulating ADH and MIC-1 to be used as diagnostic markers in Egyptian patients and assess their value either alone or combined with CA19-9 in early detection of PC. METHODS: Alcohol dehydrogenase (ADH), macrophage inhibitory cytokine (MIC-1) and CA19-9 were measured by ELISA in serum procured from PC patients (n = 50) versus normal subjects (n = 20). RESULTS: Our results demonstrate that the circulating levels of ADH, MIC-1 and CA19-9 in blood of PC were significantly higher than in healthy controls (HCs) (p < 0.001). The highest marker sensitivity observed at early stage was MIC-1 (90%) and specificity was ADH (83%). The level of all three markers was elevated significantly in early stage of PC in comparison to HCs. The addition of ADH and MIC-1 to CA19-9 significantly improved the efficacy of diagnosis (p = 0.023). CONCLUSION: Our data demonstrate that not only the combination of ADH and MIC-1 to CA19-9 can be used in early detection of PC but also can improve the overall quality of diagnosis of this lethal disease.


Asunto(s)
Alcohol Deshidrogenasa/sangre , Biomarcadores de Tumor/sangre , Factor 15 de Diferenciación de Crecimiento/sangre , Neoplasias Pancreáticas/diagnóstico , Anciano , Anciano de 80 o más Años , Antígeno CA-19-9/sangre , Estudios de Casos y Controles , Estudios Transversales , Egipto , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/sangre , Sensibilidad y Especificidad
17.
J Sex Med ; 12(7): 1533-44, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26062100

RESUMEN

INTRODUCTION: Peyronie's disease (PD) has frequently been associated with erectile dysfunction (ED) and may further compromise coitus. AIM: To investigate the efficacy of intratunical injection of genetically modified rat adipose tissue-derived stem cells (ADSCs) expressing human interferon α-2b (ADSCs-IFN) in decreasing fibrosis and restoring erectile function in a rat model of tunica albugineal fibrosis (TAF). METHODS: A total of 36 Sprague-Dawley rats (12 weeks old; 300-350 g) were randomly divided in six equal groups: (i) sham group (50 µL saline-injected into the tunica albuginea [TA]); (ii) TAF group (transforming growth factor [TGF]-ß1 [0.5 µg/50 µL] injected into the TA); (iii) TGF-ß1 plus 5 × 10(5) control ADSCs injected same day; (iv) TGF-ß1 plus 5 × 10(5) ADSCs-IFN injected same day; (v) TGF-ß1 plus 5 × 10(5) control ADSCs injected after 30 days; and (vi) TGF-ß1 plus 5 × 10(5) ADSCs-IFN injected after 30 days. Rat allogeneic ADSCs were harvested from inguinal fat tissue. MAIN OUTCOME MEASURES: Forty-five days following the TGF-ß1 injection, erectile function was assessed, and penile tissues were harvested for further evaluations. RESULTS: In the same-day injection groups, intratunical injection of ADSCs and ADSC-IFN improved erectile response observed upon stimulation of cavernous nerve compared with TAF group. Intratunical ADSC-IFN injection at day 30 improved erectile responses 3.1, 1.8, and 1.3 fold at voltages of 2.5, 5.0, and 7.0, respectively, when compared with TAF group. Furthermore, at voltages of 2.5 and 5.0, treatment on day 30 with ADSCs-IFN improved erectile responses 1.6- and 1.3-fold over treatment with ADSCs alone. Local injection of ADSCs or ADSCs-IFN reduced Peyronie's-like manifestations, and these effects might be associated with a decrease in the expression of tissue inhibitors of metalloproteinases. CONCLUSION: This study documents that transplantation of genetically modified ADSCs, with or without human IFN α-2b, attenuated Peyronie's-like changes and enhanced erectile function in a rat model of TAF.


Asunto(s)
Tejido Adiposo/trasplante , Disfunción Eréctil/terapia , Interferón-alfa/farmacología , Induración Peniana/terapia , Pene/patología , Trasplante de Células Madre , Animales , Modelos Animales de Enfermedad , Fibrosis/terapia , Humanos , Inyecciones Intralesiones , Interferón alfa-2 , Masculino , Pene/inervación , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/farmacología
18.
Future Oncol ; 11(16): 2343-50, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26260812

RESUMEN

The pre-operative diagnosis of thyroid tumors is determined by gold standard fine needle aspiration (FNA) biopsy. This has been widely accepted and offers the most cost-effective approach for evaluation of thyroid nodules. However, its diagnostic accuracy can pose a challenging scenario to surgeons. These diagnostic difficulties may subject patients to unnecessary thyroidectomies for benign thyroid nodules. Thus, additional molecular tests are needed to improve the sensitivity and specificity of FNA. The role of molecular markers is being proposed to predict the type and risk of malignancy to abate the need for diagnostic thyroidectomies. This review discusses their utility and validity in pre-operative diagnosis of thyroid nodules and how these markers can enhance the accuracy of FNA cytology.


Asunto(s)
Biomarcadores de Tumor , Cuidados Preoperatorios , Nódulo Tiroideo/diagnóstico , Nódulo Tiroideo/genética , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Biopsia con Aguja Fina , Toma de Decisiones Clínicas , Análisis Mutacional de ADN , Humanos , Inmunohistoquímica , Neoplasias de la Tiroides/diagnóstico , Neoplasias de la Tiroides/genética , Nódulo Tiroideo/cirugía
19.
J Biol Chem ; 288(3): 1458-68, 2013 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-23184953

RESUMEN

Minocycline protects against asthma independently of its antibiotic function and was recently reported as a potent poly(ADP-ribose) polymerase (PARP) inhibitor. In an animal model of asthma, a single administration of minocycline conferred excellent protection against ovalbumin-induced airway eosinophilia, mucus hypersecretion, and Th2 cytokine production (IL-4/IL-5/IL-12(p70)/IL-13/GM-CSF) and a partial protection against airway hyperresponsiveness. These effects correlated with pronounced reduction in lung and sera allergen-specific IgE. A reduction in poly(ADP-ribose) immunoreactivity in the lungs of minocycline-treated/ovalbumin-challenged mice correlated with decreased oxidative DNA damage. The effect of minocycline on PARP may be indirect, as the drug failed to efficiently block direct PARP activation in lungs of N-methyl-N'-nitro-N-nitroso-guanidine-treated mice or H(2)O(2)-treated cells. Minocycline blocked allergen-specific IgE production in B cells potentially by modulating T cell receptor (TCR)-linked IL-4 production at the mRNA level but not through a modulation of the IL-4-JAK-STAT-6 axis, IL-2 production, or NFAT1 activation. Restoration of IL-4, ex vivo, rescued IgE production by minocycline-treated/ovalbumin-stimulated B cells. IL-4 blockade correlated with a preferential inhibition of the NF-κB activation arm of TCR but not GSK3, Src, p38 MAPK, or ERK1/2. Interestingly, the drug promoted a slightly higher Src and ERK1/2 phosphorylation. Inhibition of NF-κB was linked to a complete blockade of TCR-stimulated GATA-3 expression, a pivotal transcription factor for IL-4 expression. Minocycline also reduced TNF-α-mediated NF-κB activation and expression of dependent genes. These results show a potentially broad effect of minocycline but that it may block IgE production in part by modulating TCR function, particularly by inhibiting the signaling pathway, leading to NF-κB activation, GATA-3 expression, and subsequent IL-4 production.


Asunto(s)
Asma/tratamiento farmacológico , Factor de Transcripción GATA3/genética , Factores Inmunológicos/uso terapéutico , Inflamación/tratamiento farmacológico , Interleucina-4/genética , Minociclina/uso terapéutico , FN-kappa B/genética , Receptores de Antígenos de Linfocitos T/genética , Animales , Asma/complicaciones , Asma/genética , Asma/inmunología , Factor de Transcripción GATA3/agonistas , Factor de Transcripción GATA3/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Inmunoglobulina E/genética , Inmunoglobulina E/inmunología , Factores Inmunológicos/farmacología , Inflamación/complicaciones , Inflamación/genética , Inflamación/inmunología , Interleucina-4/antagonistas & inhibidores , Interleucina-4/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Minociclina/farmacología , FN-kappa B/agonistas , FN-kappa B/inmunología , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/inmunología , Receptores de Antígenos de Linfocitos T/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal/efectos de los fármacos
20.
Mol Med ; 20: 270-9, 2014 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-24869908

RESUMEN

PDZ domain containing 1 (PDZK1) is a scaffold protein that plays a role in the fate of several proteins. Estrogen can induce PDZK1 gene expression; however, our recent report showed that PDZK1 expression in the breast cancer cell line MCF-7 is indirect and involves insulin-like growth factor (IGF)-1 receptor function. Such a relationship was established in cell culture systems and human breast cancer tissues. Here we show that overexpression of PDZK1 promoted an increase in cyclin D1 and enhanced anchorage-independent growth of MCF-7 cells in the absence of 17ß-estradiol, suggesting that PDZK1 harbors oncogenic activity. Indeed, PDKZ1 overexpression enhanced epidermal growth factor receptor (EGFR)-stimulated MEK/ERK1/2 signaling and IGF-induced Akt phosphorylation. PDZK1 appeared to play this role, in part, by stabilizing the integrity of the growth promoting factors Akt, human epidermal growth factor receptor 2 (Her2/Neu) and EGFR. Increased Akt levels occurred via a decrease in the ubiquitination of the kinase. PDZK1 overexpression was associated with resistance to paclitaxel/5-fluorouracil/etoposide only at low concentrations. Although the increased stability of Akt was sensitive to heat shock protein 90 (HSP90) inhibition, increased levels of the cochaperone cell division cycle 37 (Cdc37), as well as its ability to bind PDZK1, appear to play a larger role in kinase stability. Using human tissue microarrays, we show strong positive correlation between PDZK1, Akt and Cdc37 protein levels, and all correlated with human breast malignancy. There were no positive correlations between PDZK1 and Cdc37 at the mRNA levels, confirming our in vitro studies. These results demonstrate a relationship between PDZK1, Akt and Cdc37, and potentially Her2/Neu and EGFR, in breast cancer, representing a new axis that can be targeted therapeutically to reduce the burden of human breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Chaperoninas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proliferación Celular , Femenino , Humanos , Células MCF-7 , Proteínas de la Membrana
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA