Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Pol J Pathol ; 64(4): 308-11, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24375047

ABSTRACT

Clear cell adenocarcinoma (CCA) in the urinary tract is a rare neoplasm morphologically identical to the Müllerian counterpart. Clear cell adenocarcinoma is extremely rare in the upper urinary tract. We present a case with CCA of the renal pelvis. Microscopically, the tumor exhibited exophytic growth with predominantly tubulocystic structures, as well as solid and papillary patterns. The neoplastic cells were cuboidal with clear to pale eosinophilic cytoplasm and abundant intracellular and extracellular eosinophilic hyaline globules. By immunohistochemically, the tumor was labeled by cytokeratins and hepatocyte nuclear factor-1ß. The patient was still alive without evidence of recurrence in the follow-up period of nineteen months after diagnosis.


Subject(s)
Adenocarcinoma, Clear Cell/pathology , Biomarkers, Tumor/metabolism , Kidney Neoplasms/pathology , Abdomen/pathology , Adult , Female , Hepatocyte Nuclear Factor 1-beta/metabolism , Humans , Keratin-7/metabolism , Kidney Pelvis/pathology , Radiography, Abdominal
2.
Int J Clin Pract ; 66(2): 139-45, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22257039

ABSTRACT

AIMS: To investigate if the International Prostate Symptom Score (IPSS) voiding-to-storage subscore ratio (IPSS-V/S) can help to guide the treatment for male lower urinary tract symptoms (LUTS). METHODS: Men aged 40 years or older with a total IPSS (IPSS-T) 8 or more were constitutively enrolled from January 2010 to December 2010. The IPSS voiding (IPSS-V) and storage subscore (IPSS-S) were recorded separately, and the IPSS-V/S was calculated. Patients were divided into two groups according to the baseline IPSS-V/S value. First-line doxazosin (4 mg per day) and tolterodine (4 mg per day) monotherapy were given to patients with IPSS-V/S > 1 and IPSS-V/S ≤ 1, respectively. The IPSS-T, IPSS-V, IPSS-S, quality of life (QoL), maximum flow rate (Qmax), voided volume and postvoid residual (PVR) were measured at 1 month (visit 1) and 3 months (visit 2) after treatment. RESULTS: After medical treatment for 1 month, 89/116 (76.7%) patients receiving tolterodine and 218/279 (78.1%) patients receiving doxazosin reported an improved outcome (global response assessment, GRA ≥ 1 point). The mean IPSS-T, IPSS-S decreased, and QoL improved significantly in both groups. Significant increased Qmax, voided volume, decreased IPSS-V and PVR were noted only in patients receiving doxazosin. There was no significant increase of PVR (from 50.1 to 60.4 ml, p = 0.106), and no patient developed urinary retention after tolterodinie monotherapy for 1 month. However, patients aged more than 70 years had significant association with increased PVR (≥ 50 ml). CONCLUSION: Initial treatment with doxazosin for patients with IPSS-V/S > 1 and tolterodine for patients with IPSS-V/S ≤ 1 is safe and feasible. Elderly people (≥ 70 years) and patients with Qmax < 10 ml/s are more likely to have increased PVR (≥ 50 ml).


Subject(s)
Adrenergic alpha-1 Receptor Antagonists/therapeutic use , Benzhydryl Compounds/therapeutic use , Cresols/therapeutic use , Doxazosin/therapeutic use , Muscarinic Antagonists/therapeutic use , Phenylpropanolamine/therapeutic use , Prostatism/drug therapy , Aged , Feasibility Studies , Humans , Male , Middle Aged , Prospective Studies , Severity of Illness Index , Tolterodine Tartrate , Treatment Outcome
3.
Anaesthesia ; 66(9): 791-5, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21707563

ABSTRACT

Increased tracheal cuff pressure during mechanical ventilation is associated with reduced mucosal blood flow and ischaemia, as well as postoperative sore throat. We assessed the potential effects of transoesophageal echocardiography probe insertion on the tracheal cuff pressure in patients undergoing cardiac surgery. Using a manometer, the cuff pressure of a high-volume, low-pressure tracheal tube (inner diameter 7.0 mm for women and 7.5 mm for men) was adjusted to 25-30 cm H(2)O before blind insertion of a transoesophageal echocardiography probe. The pressure changes were then recorded for 1 min. After probe insertion, the mean (SD) intra-cuff pressure increased from 27.7 (1.5) to 36.2 (6.4) cm H(2)O (p < 0.001) and was > 35 cm H(2)0 in 17/38 patients (45%). Our results suggest that transoesophageal echocardiography probe insertion may increase the tracheal cuff pressure more than that is generally recommended and therefore the latter should be routinely monitored under such circumstances.


Subject(s)
Cardiac Surgical Procedures , Echocardiography, Transesophageal/adverse effects , Intubation, Intratracheal/instrumentation , Adult , Aged , Echocardiography, Transesophageal/instrumentation , Female , Humans , Male , Middle Aged , Pressure , Trachea/blood supply
4.
Mar Biotechnol (NY) ; 23(2): 201-214, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33580373

ABSTRACT

Sexual dimorphism is common across the animal kingdom. Knowledge of the mechanisms of sexual size dimorphism is limited although it is important in biology and aquaculture. Tilapia is the common name for ~ 100 species of cichlid fish. Some are important aquaculture species and males outgrow females. To gain novel insights into the mechanisms underlying sexual size dimorphism, we analyzed the differences of brain transcriptomes between males and females in Mozambique tilapia and studied the function of the pro-opiomelanocortin (Pomc) gene in tilapia and zebrafish. The transcriptome analysis identified 123, 55, and 2706 sex-biased genes at 5, 30, and 90 dph (days post-hatch), respectively, indicating sexual dimorphism of gene expressions in the brain. The expression of Pomc in the tilapia brain was a female-biased at 30, 90, and 120 dph. An analysis of the DNA sequence located upstream of the tilapia Pomc transcriptional start site identified two estrogenic response elements. In vitro luciferase assay of the two elements revealed that ß-estradiol significantly enhanced the expression of luciferase activity, suggesting that the expression of Pomc is mediated by estrogen. We knocked out Pomc in zebrafish using Crispr/Cas-9. The Pomc-knockout zebrafish showed faster growth and higher sensitivity to feeding as compared to the wild-type fish. Taken together, our results indicate that Pomc contributes to sexual size dimorphism and suggest that the high estrogen level in females promotes the expression of Pomc and suppresses feeding in female tilapias, which leads to the slower growth of female tilapias.


Subject(s)
Pro-Opiomelanocortin/genetics , Sex Characteristics , Tilapia/genetics , Zebrafish/genetics , Animals , Brain/metabolism , Estrogens , Feeding Behavior/drug effects , Female , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Male , Sequence Analysis, DNA , Tilapia/growth & development , Zebrafish/growth & development
5.
Anaesthesia ; 65(1): 4-7, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19889113

ABSTRACT

We performed a randomised, double-blind study to evaluate the effect of the pre-emptive use of minimal dose intravenous fentanyl (25 microg) on the incidence of cough caused by a larger bolus of intravenous fentanyl. Six hundred patients were randomly assigned to one of three groups to receive either 0.5 ml saline 0.9% 1 min before administration of fentanyl 150 microg (3 ml), or pre-emptive fentanyl 25 microg (0.5 ml) 1 min before administration of fentanyl 125 microg or 150 microg. The incidence of fentanyl-induced cough was significantly lower in both pre-emptive groups (7 (3.5%) for 125 microgfentanyl and 15 (7.5%) for 150 microg fentanyl) than in the saline group (37 (18.5%); p = 0.001). We conclude that pre-emptive use of fentanyl 25 microg, administered 1 min before bolus injection of fentanyl (125 or 150 microg), can effectively suppress fentanyl-induced cough.


Subject(s)
Anesthetics, Intravenous/administration & dosage , Cough/prevention & control , Fentanyl/administration & dosage , Intraoperative Complications/prevention & control , Adolescent , Adult , Aged , Anesthetics, Intravenous/adverse effects , Blood Pressure/drug effects , Cough/chemically induced , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Female , Fentanyl/adverse effects , Heart Rate/drug effects , Humans , Injections, Intravenous , Intraoperative Complications/chemically induced , Male , Middle Aged , Young Adult
6.
Oncogene ; 37(4): 461-477, 2018 01 25.
Article in English | MEDLINE | ID: mdl-28967906

ABSTRACT

Recently, we reported that the histone methyltransferase, EZH2, controls leukocyte migration through interaction with the cytoskeleton remodeling effector, VAV, and direct methylation of the cytoskeletal regulatory protein, Talin. However, it is unclear whether this extranuclear, epigenetic-independent function of EZH2 has a profound impact on the initiation of cellular transformation and metastasis. Here, we show that EZH2 increases Talin1 methylation and cleavage, thereby enhancing adhesion turnover and promoting accelerated tumorigenesis. This transforming capacity is abolished by targeted disruption of EZH2 interaction with VAV. Furthermore, our studies demonstrate that EZH2 in the cytoplasm is closely associated with cancer stem cell properties, and that overexpression of EZH2, a mutant EZH2 lacking its nuclear localization signal (EZH2ΔNLS), or a methyl-mimicking Talin1 mutant substantially promotes JAK2-dependent STAT3 activation and cellular transformation. Taken together, our results suggest a critical role for the VAV interaction-dependent, extranuclear action of EZH2 in neoplastic transformation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Regulation, Neoplastic , Neoplasms/pathology , Proto-Oncogene Proteins c-vav/metabolism , Animals , Cell Adhesion/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/pathology , Cytoplasm/genetics , Cytoplasm/pathology , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/isolation & purification , Female , Gene Knockdown Techniques , HEK293 Cells , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Jurkat Cells , Methylation , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Mutagenesis, Site-Directed , Neoplasms/genetics , Nuclear Localization Signals/genetics , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Talin/genetics , Talin/metabolism , Xenograft Model Antitumor Assays
7.
Clin Cancer Res ; 7(9): 2880-6, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11555606

ABSTRACT

One of the potential therapeutic interventions to hormone-independent breast cancer would be to reactivate the expression of estrogen receptor or progesterone receptor (PR) in the tumor cells so as to render the tumor responsive to the hormones. We have reported previously that progesterone markedly inhibited cell growth and induced remarkable focal adhesions in PR-transfected MDA-MB-231 cells. The aim of this study was to determine the effects of progesterone on the invasive properties and in vivo tumor growth of PR-transfected MDA-MB-231 cells. It was found that progesterone has increased cell resistance to trypsin digestion and increased cell attachment to extracellular matrix proteins, especially laminin and fibronectin. In vitro invasion assays using modified Boyden chambers showed that progesterone increased cell migration through matrix protein-coated membranes. However, Northern blotting analysis demonstrated that progesterone strongly down-regulated (up to 60-fold) the gene expression of urokinase plasminogen activator and increased (up to 5-fold) the expression of tissue-type plasminogen activator in these cells. This pattern of gene regulation suggested an inhibition of cell invasiveness because numerous clinical studies have indicated that low levels of urokinase plasminogen activator and high levels of tissue-type plasminogen activator in breast cancer are associated with favorable prognosis. Furthermore, animal studies showed that progesterone strongly inhibited the tumor formation and growth in Scid mice. After 12 weeks of inoculation, the median weight of tumors in the progesterone-treated group was 25 mg compared with 203 mg in the placebo group (P < 0.001). These results suggest that progesterone may provide effective treatment for estrogen receptor- and PR-negative breast cancer if the PR expression were reactivated. Alternatively, activation of progesterone-mediated molecular pathways in hormone-independent breast cancer may achieve similar therapeutic effects.


Subject(s)
Cell Division/drug effects , Progesterone/pharmacology , Receptors, Progesterone/physiology , Animals , Cell Adhesion/drug effects , Cell Movement/drug effects , Dose-Response Relationship, Drug , Extracellular Matrix Proteins/pharmacology , Female , Humans , Mice , Mice, SCID , Neoplasm Invasiveness , Neoplasm Transplantation , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Progesterone/genetics , Time Factors , Tissue Plasminogen Activator/genetics , Transfection , Trypsin/pharmacology , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/genetics , Xenograft Model Antitumor Assays
8.
Clin Cancer Res ; 5(2): 395-403, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10037189

ABSTRACT

Because progesterone exerts its effects mainly via estrogen-dependent progesterone receptor (PgR), the expression of progesterone's effects may be overshadowed by the priming effect of estrogen. PgR expression vectors were transfected into estrogen receptor (ER)-alpha and PgR-negative breast cancer cells MDA-MB-231; thus the functions of progesterone could be studied independent of estrogens and ERs. Eight stable transfectant clones expressing both PgR isoform A and B were studied for their growth response to progesterone and its analogues. Although progesterone had no effect on growth in the control transfectant, the hormone markedly inhibited DNA synthesis and cell growth in all of the PgR-transfectants dose-dependently from 10(-12)-10(-6) M. This growth inhibition was associated with an arrest of cells in the G0/G1 phase of the cell cycle. Progestins medroxyprogesterone acetate, Org2058, and R5020 also strongly inhibited DNA synthesis, and their doses required for maximal inhibition of 60-70% were 10(-17) M, 10(-13) M, and 10(-7) M, respectively. Antiprogestin ZK98299 alone had no effect, but the compound was capable of counteracting the inhibitory effect of progesterone. In contrast, RU486 inhibited DNA synthesis, and it showed no further effects when it was used concurrently with progesterone. These results indicate that progestins are per se antiproliferative via a PgR-mediated mechanism in breast cancer cells. More importantly, we have shown that progestins may exert effective inhibitory control over the cell growth if the PgR expression is reactivated in ER- and PgR-negative breast cancer cells.


Subject(s)
Cell Division/drug effects , Progestins/pharmacology , Receptors, Progesterone/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Cycle/drug effects , DNA/biosynthesis , DNA/drug effects , DNA, Complementary , Estradiol/pharmacology , Humans , Progesterone/pharmacology , Progestins/antagonists & inhibitors , Receptors, Estrogen/metabolism , Receptors, Progesterone/genetics , Transfection , Tumor Cells, Cultured
9.
Mol Endocrinol ; 14(3): 348-58, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10707953

ABSTRACT

Since the effects of progesterone are mediated mainly via estrogen-dependent progesterone receptor (PR), the expression of the effects of progesterone may be masked or overridden by the influence of estrogen under conditions in which priming with estrogens is required. We have established a PR-positive but estrogen receptor-alpha (ER-alpha) negative breast cancer cell model by transfecting PR cDNA into ER-alpha- and PR-negative MDA-MB-231 cells in order that the functions of progesterone can be studied independently of estrogens. We have demonstrated using this model that progesterone markedly inhibited cell growth. We have also discovered that progesterone induced remarkable changes in cell morphology and specific adhesion structures. Progesterone-treated cells became considerably more flattened and well spread than vehicle-treated control cells. This was associated with a striking increase of stress fibers, both in number and diameter, and increased focal contacts as shown by the staining of focal adhesion proteins paxillin and talin. There were also distinct increases in tyrosine phosphorylation of focal adhesion protein paxillin and focal adhesion kinase in association with increased focal adhesion. The staining of tyrosine-phosphorylated proteins was concentrated at focal adhesions in progesterone-treated cells. More interestingly, monoclonal antibody (Ab) to beta1 integrin was able to inhibit progesterone-induced cell spreading and formation of actin cytoskeleton. To our knowledge, this is the first report describing a direct effect of progesterone in inducing spreading and adhesion of breast cancer cells, and beta1-integrin appeared to play an essential role in the effect. It is known that the initial step of tumor metastasis is the breakaway of tumor cells from primary tumor mass when they lose the ability to attach. Hence, progesterone-induced cell spreading and adhesion may have significant implications in tumor metastasis.


Subject(s)
Breast Neoplasms/pathology , Carcinoma/pathology , Cell Adhesion/drug effects , Progesterone/pharmacology , Receptors, Progesterone/drug effects , Breast Neoplasms/metabolism , Carcinoma/metabolism , Cell Adhesion Molecules/metabolism , Cell Size , Cytoskeletal Proteins/metabolism , DNA, Complementary/genetics , Estrogen Receptor alpha , Estrogen Receptor beta , Female , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans , Integrin beta1/physiology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Paxillin , Phosphoproteins/metabolism , Phosphorylation , Protein Processing, Post-Translational , Protein-Tyrosine Kinases/metabolism , Receptors, Estrogen/deficiency , Receptors, Estrogen/physiology , Receptors, Progesterone/genetics , Receptors, Progesterone/physiology , Recombinant Fusion Proteins/physiology , Signal Transduction , Transfection , Tumor Cells, Cultured/drug effects
10.
Br J Cancer ; 83(3): 319-23, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10917545

ABSTRACT

Metallothioneins (MTs), a group of ubiquitous metalloproteins, comprise isoforms encoded by ten functional genes in humans. Different MT isoforms possibly play different functional roles during development or under various physiological conditions. The MT-1E isoform mRNA has been recently shown to be differentially expressed in oestrogen receptor (OR)-positive and OR-negative breast cancer cell lines. In this study, we evaluated MT-1E mRNA expression via semi-quantitative RT-PCR in 51 primary invasive ductal breast cancer tissues, concurrently with OR-positive and progesterone receptor (PR)-positive MCF7 cells, OR-negative and PR-negative MDA-MB-231 cells and PR-transfected MDA-MB-231 breast cancer cells (ABC28). We demonstrated significantly higher MT-1E mRNA expression in OR-negative compared with OR-positive breast cancer tissues (P = 0.026). MCF7 cells lacked MT-1E mRNA expression, while both OR- and PR-negative MDA-MD-231 cells exhibited a high level of MT-1E mRNA expression. The level of MT-1E mRNA expression in progesterone-treated and -untreated ABC28 cells remained similar as the parental cell line MDA-MB-231-C2 cells. The results suggest that MT-1E may have specific and functional roles in OR-negative invasive ductal breast cancers, possibly mediated via effector genes downstream of the oestrogen receptor, but not through the PR pathway.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Metallothionein/metabolism , RNA, Messenger/metabolism , Biomarkers, Tumor/metabolism , DNA Restriction Enzymes/metabolism , Electrophoresis, Agar Gel , Female , Gene Expression Regulation, Neoplastic , Genetic Markers/genetics , Humans , Immunohistochemistry , Isomerism , Metallothionein/genetics , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Transfection , Tumor Cells, Cultured
11.
Br J Cancer ; 85(12): 1978-86, 2001 Dec 14.
Article in English | MEDLINE | ID: mdl-11747343

ABSTRACT

Progesterone antagonist RU486 (mifepristone) has been implicated for many anti-neoplastic and obstetrical applications. But the compound has demonstrated undesired agonist-like effect depending on cell, tissue and species studied. Using PR-transfected breast cancer cells MDA-MB-231, this report describes the similarities and differences between progesterone- and RU486-mediated effects on cell growth, cell differentiation and, at the molecular level, on the activation of p44/p42 MAP kinases (MAPK). Like progesterone, RU486 inhibited cells growth by arresting the cells in G0/G1 phase of the cell cycle. In contrast to progesterone that induced cell spreading, RU486 induced a multipolar, stellate morphology. RU486-treated cells showed no increase of stress fibers, nor was there any increase of focal adhesions as progesterone-treated cells did. Furthermore, despite of the fact that both compounds inhibited cell growth, RU486 significantly stimulated the activation of p44/p42 MAP kinases whereas progesterone markedly inhibited the activation. Nonetheless, the effects of RU486 were PR-mediated and RU486 was able to antagonize the effect of progesterone on cell growth and focal adhesion. In conclusion, RU486 can act not only as a progesterone antagonist, a progesterone agonist but also induced morphological and molecular changes that were distinct from progesterone-mediated effects in PR-transfected MDA-MB-231 cells. The non-progesterone-like effect of RU486 may be mediated through a pathway that is different from the progesterone-mediated pathway, or it is the result of a blockade of certain critical step(s) in the progesterone-mediated pathway. In any case, undesired side effects of antiprogestin may create clinical complications. PR-transfected MDA-MB-231 breast cancer cells provide a model for studying the functions of progesterone analogues.


Subject(s)
Breast Neoplasms/pathology , Hormone Antagonists/pharmacology , Mifepristone/pharmacology , Progesterone/pharmacology , Receptors, Progesterone/drug effects , Cell Cycle/drug effects , Cell Division/drug effects , Cell Size/drug effects , Enzyme Activation/drug effects , Female , Focal Adhesions/drug effects , Gonanes/pharmacology , Humans , MAP Kinase Signaling System/drug effects , Progesterone/administration & dosage , Receptors, Progesterone/agonists , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/genetics , Recombinant Fusion Proteins/drug effects , Stress Fibers/ultrastructure , Transfection , Tumor Cells, Cultured/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL