Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Foot Ankle Surg ; 61(4): 735-738, 2022.
Article in English | MEDLINE | ID: mdl-34953668

ABSTRACT

The standard management for residual osteomyelitis following amputation in a diabetic foot infection includes a prolonged course of intravenous antibiotic agents. The purpose of this analysis was to investigate whether oral antibiotic therapy led to treatment failure more than intravenous antibiotic therapy for this indication. The primary endpoint was treatment failure within one year of the initial amputation, defined by re-operation for residual osteomyelitic infection or a remaining nonhealing wound at the surgical site. All patients received at least 4 weeks of antibiotics and were chosen for oral or intravenous route of administration by infectious disease specialists. Characteristics including age, sex, hemoglobin A1c, BMI, tobacco use, PVD, homelessness and IDSA classification were also assessed for influence on antibiotic success and failure. Of the 65 patients meeting inclusion criteria, treatment failure occurred in 32 and treatment success occurred in 33. Of the treatment failures, 17 (53%) were in the intravenous group, and 15 (47%) were in the oral group. The differences between the modalities of antibiotic administration and their failure rates were not found to be statistically significant (p = .28 (proportional difference: -14%, 95% confidence interval [CI]:-36% to 10%)).


Subject(s)
Diabetes Mellitus , Diabetic Foot , Osteomyelitis , Administration, Oral , Amputation, Surgical , Anti-Bacterial Agents , Diabetes Mellitus/drug therapy , Diabetic Foot/drug therapy , Diabetic Foot/surgery , Humans , Osteomyelitis/drug therapy , Osteomyelitis/etiology , Osteomyelitis/surgery
3.
Oncotarget ; 8(24): 39087-39100, 2017 Jun 13.
Article in English | MEDLINE | ID: mdl-28388569

ABSTRACT

Cytochrome P450 1B1 (CYP1B1) is recognized as a universal tumor biomarker and a feasible therapeutic target due to its specific overexpression in cancer tissues. Despite its up-regulation in prostate cancer (PCa), biological significance and clinicopathological features of CYP1B1 are still elusive. Here, we show that overexpression or hyperactivation of CYP1B1 stimulated proliferative, migratory and invasive potential of non-tumorigenic PCa cells. Attenuation of CYP1B1 with its specific small hairpin (sh) RNAs greatly reduced proliferation through apoptotic cell death and impaired migration and invasion in PCa cells. Intratumoral injection of CYP1B1 shRNA attenuated growth of pre-existing tumors. The antitumor effect of CYP1B1 shRNA was also observed in prostate tumor xenograft mouse models. Among the genes altered by CYP1B1 knockdown, reduction of caspase-1 (CASP1) activity attenuated the antitumor effect of CYP1B1 inhibition. Indeed, CYP1B1 regulates CASP1 expression or activity. Finally, CYP1B1 expression was increased in higher grades of PCa and overall survival was significantly reduced in patients with high levels of CYP1B1 protein. CYP1B1 expression was reversely associated with CASP1 expression in clinical tissue samples. Together, our results demonstrate that CYP1B1 regulates PCa tumorigenesis by inhibiting CASP1 activation. Thus, the CYP1B1-CASP1 axis may be useful as a potential biomarker and a therapeutic target for PCa.


Subject(s)
Biomarkers, Tumor/metabolism , Caspase 1/metabolism , Cell Transformation, Neoplastic/pathology , Cytochrome P-450 CYP1B1/metabolism , Prostatic Neoplasms/pathology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Caspase 1/genetics , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cytochrome P-450 CYP1B1/antagonists & inhibitors , Cytochrome P-450 CYP1B1/genetics , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Prognosis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA, Small Interfering/genetics , Survival Rate , Tumor Cells, Cultured , Up-Regulation , Xenograft Model Antitumor Assays
4.
Oncotarget ; 6(18): 16341-51, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-26036629

ABSTRACT

DNA mismatch repair (MMR) enzymes act as proofreading complexes that maintains genomic integrity and MMR-deficient cells show an increased mutation rate. MMR has also been shown to influence cell signaling and the regulation of tumor development. MMR consists of various genes and includes post-meiotic segregation (PMS) 2 which is a vital component of mutL-alpha. In prostate, the functional role of this gene has never been reported and in this study, our aim was to investigate the effect of PMS2 on growth properties of prostate cancer (PCa) cells. Previous studies have shown PMS2 to be deficient in DU145 cells and this lack of expression was confirmed by Western blotting whereas normal prostatic PWR-1E and RWPE-1 cells expressed this gene. PMS2 effects on various growth properties of DU145 were then determined by creating stable gene transfectants. Interestingly, PMS2 caused decreased cell proliferation, migration, invasion, and in vivo growth; and increased apoptosis as compared to vector control. We further analyzed genes affected by PMS2 expression and observe the apoptosis-related TMS1 gene to be significantly upregulated whereas anti-apoptotic BCL2A1 was downregulated. These results demonstrate a functional role for PMS2 to protect against PCa progression by enhancing apoptosis of PCa cells.


Subject(s)
Adenosine Triphosphatases/metabolism , Apoptosis/genetics , DNA Mismatch Repair/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Adenosine Triphosphatases/genetics , CARD Signaling Adaptor Proteins , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cytoskeletal Proteins/biosynthesis , DNA/genetics , DNA Repair Enzymes/genetics , DNA-Binding Proteins/genetics , Humans , Male , Minor Histocompatibility Antigens , Mismatch Repair Endonuclease PMS2 , Neoplasm Invasiveness/genetics , Prostate , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , RNA Interference , RNA, Small Interfering
5.
Oncotarget ; 6(10): 7774-87, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25860934

ABSTRACT

Despite high protein expression and enzymatic activity of cytochrome P450 1B1 (CYP1B1) in renal cell cancer (RCC), its functional significance has not been elucidated. Here we explored the functional role and regulatory mechanism of CYP1B1 in RCC. Reduction of CYP1B1 levels fail to prevent in vitro tumorigenicity such as proliferation, apoptosis, and cell cycle progression of RCC cells. Moreover, the expression levels are not associated with tumor type, stage, Fuhrman grade and 5-year survival probability after surgery. Instead, alteration of CYP1B1 expression regulates the chemosensitivity of RCC cells to docetaxel suggesting its critical contribution to the chemoresistance. Additionally, miR-200c, which is significantly down-regulated in RCC regulates CYP1B1 expression and activity. An inverse association was also observed between the expression levels of miR-200c and CYP1B1 protein in RCC tissues. Finally, alteration of miR-200c levels affects the chemosensitivity of RCC cells. Restoration of docetaxel resistance by exogenous expression of CYP1B1 in miR-200c-over-expressing cells indicates that CYP1B1 is a functional target of miR-200c. These results suggest that CYP1B1 up-regulation mediated by low miR-200c is one of the mechanisms underlying resistance of RCC cells to docetaxel. Therefore, expression of CYP1B1 and miR-200c in RCC may be useful as a prediction for docetaxel response.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Cytochrome P-450 CYP1B1/metabolism , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , MicroRNAs/genetics , Taxoids/pharmacology , Aged , Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cytochrome P-450 CYP1B1/genetics , Docetaxel , Drug Resistance, Neoplasm , Female , Humans , Kidney Neoplasms/pathology , Male , MicroRNAs/metabolism , Middle Aged , Up-Regulation
6.
Oncotarget ; 5(22): 11297-307, 2014 Nov 30.
Article in English | MEDLINE | ID: mdl-25526032

ABSTRACT

Mismatch repair (MMR) enzymes have been shown to be deficient in prostate cancer (PCa). MMR can influence the regulation of tumor development in various cancers but their role on PCa has not been investigated. The aim of the present study was to determine the functional effects of the mutL-homolog 1 (MLH1) gene on growth of PCa cells. The DU145 cell line has been established as MLH1-deficient and thus, this cell line was utilized to determine effects of MLH1 by gene expression. Lack of MLH1 protein expression was confirmed by Western blotting in DU145 cells whereas levels were high in normal PWR-1E and RWPE-1 prostatic cells. MLH1-expressing stable transfectant DU145 cells were then created to characterize the effects this MMR gene has on various growth properties. Expression of MLH1 resulted in decreased cell proliferation, migration and invasion properties. Lack of cell growth in vivo also indicated a tumor suppressive effect by MLH1. Interestingly, MLH1 caused an increase in apoptosis along with phosphorylated c-Abl, and treatment with MLH1 siRNAs countered this effect. Furthermore, inhibition of c-Abl with STI571 also abrogated the effect on apoptosis caused by MLH1. These results demonstrate MLH1 protects against PCa development by inducing c-Abl-mediated apoptosis.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , DNA Mismatch Repair , Nuclear Proteins/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Adaptor Proteins, Signal Transducing/biosynthesis , Animals , Apoptosis/genetics , Benzamides/pharmacology , Cell Line, Tumor , Heterografts , Humans , Imatinib Mesylate , Male , Mice , Mice, Nude , MutL Protein Homolog 1 , Nuclear Proteins/biosynthesis , Phosphorylation , Piperazines/pharmacology , Prostatic Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/metabolism , Pyrimidines/pharmacology , Transfection
7.
Tumour Biol ; 35(10): 10223-30, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25027399

ABSTRACT

The cytochrome P450 1B1 (CYP1B1) enzyme activates xenobiotics to reactive forms as well as convert estradiol to 4-hydroxy-estradiol that has been shown to play a role in the carcinogenesis process of the kidney in male but not female animals. Prior reports show polymorphic variants of CYP1B1 to alter catalytic activity, and thus, we hypothesize that polymorphisms of the CYP1B1 gene are involved in the malignant transformation of the renal cell in men. The genetic distributions of five CYP1B1 polymorphisms were analyzed by polymerase chain reaction-restriction fragment length polymorphism in 480 normal healthy subjects and 403 sporadic renal cell carcinoma cases. All subjects were Caucasian men. The sites evaluated were codons 48 (C → G, Arg → Gly, rs10012), 119 (G → T, Ala → Ser, rs1056827), 432 (C → G, Leu → Val, rs1056836), 449 (C → T, Asp, rs1056837), and 453 (A → G, Asn → Ser, rs1800440). A trend was demonstrated for the 432 Val/Val (χ2, P = 0.06) and 449 T/T (χ2, P = 0.1) genotypes to play a protective role against renal cancer. Odds ratio (95 % confidence interval) for Val/Val compared to Leu/Leu at codon 432 was 0.65 (0.44-0.95) and T/T compared to C/C at codon 449 was 0.67 (0.45-0.99). Codons 432 and 449 were observed to be linked (D = 0.24), and haplotype involving 432 Val and 449 T was significantly reduced in cancer cases (P = 0.04). No association was found, however, when analyzing polymorphic sites with clinical stage of cancer. These results demonstrate polymorphisms of CYP1B1 to be associated with renal carcinogenesis and are of importance in understanding their role in the pathogenesis of renal cell carcinoma.


Subject(s)
Carcinoma, Renal Cell/genetics , Cytochrome P-450 CYP1B1/genetics , Genetic Predisposition to Disease/genetics , Kidney Neoplasms/genetics , Polymorphism, Single Nucleotide/genetics , Adult , Aged , Aged, 80 and over , Carcinoma, Renal Cell/enzymology , Genotype , Humans , Kidney Neoplasms/enzymology , Male , Middle Aged , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...