Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
BMC Cancer ; 24(1): 397, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38553680

ABSTRACT

BACKGROUND: High-risk stage III colon cancer has a considerably poorer prognosis than stage II and low-risk stage III colon cancers. Nevertheless, most guidelines recommend similar adjuvant treatment approaches for all these stages despite the dearth of research focusing on high-risk stage III colon cancer and the potential for improved prognosis with intensive adjuvant treatment. Given the the proven efficacy of triplet chemotherapy in metastatic colorectal cancer treatment, the goal of this study is to evaluate the oncologic efficacy and safety of mFOLFIRINOX in comparison to those of the current standard of care, mFOLFOX 6, as an adjuvant treatment for patients diagnosed with high-risk stage III colon cancer after radical resection. METHODS: This multicenter, randomized (1:1), open-label, phase II trial will assess and compare the effectiveness and toxicity of mFOLFIRINOX and mFOLFOX 6 in patients with high-risk stage III colon cancer after radical resection. The goal of the trial is to enroll 312 eligible patients, from 11 institutes, aged between 20 and 70 years, with an Eastern Cooperative Oncology Group (ECOG) performance status of 0-2, or between 70 and 75 with an ECOG performance status of 0. Patients will be randomized into two arms - Arm A, the experimental arm, and Arm B, the reference arm - and will receive 12 cycles of mFOLFIRINOX and mFOLFOX 6 every 2 weeks, respectively. The primary endpoint of this study is the 3-year disease-free survival, and secondary endpoints include the 3-year overall survival and treatment toxicity. DISCUSSION: The Frost trial would help determine the oncologic efficacy and safety of adjuvant triplet chemotherapy for high-risk stage III colon cancers and ultimately improve prognoses. TRIAL REGISTRATION: ClinicalTrials.gov NCT05179889, registered on 17 December 2021.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Colonic Neoplasms , Adult , Aged , Humans , Middle Aged , Young Adult , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Chemotherapy, Adjuvant , Clinical Trials, Phase II as Topic , Colonic Neoplasms/pathology , Disease-Free Survival , Multicenter Studies as Topic , Progression-Free Survival , Randomized Controlled Trials as Topic , Fluorouracil/therapeutic use
2.
Biochem Biophys Res Commun ; 503(4): 2639-2645, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30107910

ABSTRACT

Coenzyme Q10 (CoQ10) protects retinal ganglion cells (RGCs) in experimental retinal ischemia and glaucoma by scavenging reactive oxygen species. We tested whether a diet supplemented with ubiquinol, the reduced form of CoQ10, promotes RGC survival and blocks the apoptotic pathway in ischemic mouse retina induced by acute high intraocular pressure (IOP) elevation. Ubiquinol (1%) treatment significantly promoted RGC survival at 2 weeks after ischemia/reperfusion. The ubiquinol treatment significantly blocked activation of astroglial and microglial cells in the ischemic retina at 2 weeks. While the ubiquinol treatment significantly decreased active Bax protein expression in the ischemic retina, phosphorylation of Bad at serine 112 and Bcl-xL protein expression were preserved in the ubiquinol-treated ischemic retina at 12 h. Consistently, the ubiquinol treatment prevented apoptotic cell death by blocking caspase-3 cleavage. These results suggest that the ubiquinol enhances RGC survival by modulating the Bax/Bad/Bcl-xL-mediated apoptotic pathway in the ischemic retina. Ubiquinol has therapeutic potential for ameliorating elevated IOP-induced ischemic retinal degeneration.


Subject(s)
Antioxidants/pharmacology , Gene Expression Regulation/drug effects , Reperfusion Injury/drug therapy , Retinal Degeneration/prevention & control , Retinal Ganglion Cells/drug effects , Ubiquinone/analogs & derivatives , Animals , Apoptosis , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Caspase 3/genetics , Caspase 3/metabolism , Cell Survival/drug effects , Disease Models, Animal , Female , Intraocular Pressure , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Oxidative Stress/drug effects , Phosphorylation/drug effects , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Signal Transduction , Ubiquinone/pharmacology , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , bcl-Associated Death Protein/genetics , bcl-Associated Death Protein/metabolism , bcl-X Protein/genetics , bcl-X Protein/metabolism
3.
Blood Res ; 51(4): 268-273, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28090490

ABSTRACT

BACKGROUND: In Korea, the prevalence of anemia and iron deficiency anemia (IDA) among older infants and young children remains high. To detect IDA early and to reduce its adverse impact, we assessed the characteristics of infants and young children who had IDA or were at risk of developing IDA, or who exhibited characteristics associated with severe anemia. METHODS: Among the 1,782 IDA-affected children aged 6 months to 18 years who visited the hospital, we retrospectively analyzed the medical records and laboratory data of 1,330 IDA-affected children aged 6-23 months who were diagnosed between 1996 and 2013. We excluded patients with a C-reactive protein level ≥5 mg/dL. RESULTS: IDA was predominant in boys (2.14:1) during infancy and early childhood. The peak IDA incidence was noted among infants aged 9-12 months. Only 7% patients exhibited symptoms of IDA, while 23.6% patients with severe IDA demonstrated classic symptoms/signs of IDA. Low birth weight (LBW) infants with IDA demonstrated low adherence to iron supplementation. In a multivariate analysis, prolonged breastfeeding without iron fortification (odds ratio [OR] 5.70), and a LBW (OR 6.49) were identified as risk factors of severe anemia. CONCLUSION: LBW infants need more attention in order to increase their adherence to iron supplementation. For the early detection of IDA, nutritional status of all infants, and iron batteries of high-risk infants (LBW infants, infants with prolonged breastfeeding, picky eaters, and/or infants with the presence of IDA symptoms) should be evaluated at their health screening visits.

4.
Invest Ophthalmol Vis Sci ; 55(2): 993-1005, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24458150

ABSTRACT

PURPOSE: To test whether a diet supplemented with coenzyme Q10 (CoQ10) ameliorates glutamate excitotoxicity and oxidative stress-mediated retinal ganglion cell (RGC) degeneration by preventing mitochondrial alterations in the retina of glaucomatous DBA/2J mice. METHODS: Preglaucomatous DBA/2J and age-matched control DBA/2J-Gpnmb(+) mice were fed with CoQ10 (1%) or a control diet daily for 6 months. The RGC survival and axon preservation were measured by Brn3a and neurofilament immunohistochemistry and by conventional transmission electron microscopy. Glial fibrillary acidic protein (GFAP), superoxide dismutase-2 (SOD2), heme oxygenase-1 (HO1), N-methyl-d-aspartate receptor (NR) 1 and 2A, and Bax and phosphorylated Bad (pBad) protein expression was measured by Western blot analysis. Apoptotic cell death was assessed by TUNEL staining. Mitochondrial DNA (mtDNA) content and mitochondrial transcription factor A (Tfam)/oxidative phosphorylation (OXPHOS) complex IV protein expression were measured by real-time PCR and Western blot analysis. RESULTS: Coenzyme Q10 promoted RGC survival by approximately 29% and preserved the axons in the optic nerve head (ONH), as well as inhibited astroglial activation by decreasing GFAP expression in the retina and ONH of glaucomatous DBA/2J mice. Intriguingly, CoQ10 significantly blocked the upregulation of NR1 and NR2A, as well as of SOD2 and HO1 protein expression in the retina of glaucomatous DBA/2J mice. In addition, CoQ10 significantly prevented apoptotic cell death by decreasing Bax protein expression or by increasing pBad protein expression. More importantly, CoQ10 preserved mtDNA content and Tfam/OXPHOS complex IV protein expression in the retina of glaucomatous DBA/2J mice. CONCLUSIONS: Our findings suggest that CoQ10 may be a promising therapeutic strategy for ameliorating glutamate excitotoxicity and oxidative stress in glaucomatous neurodegeneration.


Subject(s)
Disease Models, Animal , Glaucoma/drug therapy , Glutamic Acid/metabolism , Mitochondria/drug effects , Oxidative Stress/drug effects , Ubiquinone/analogs & derivatives , Vitamins/therapeutic use , Animals , Axons/drug effects , Axons/pathology , Blotting, Western , Female , Glaucoma/metabolism , Glial Fibrillary Acidic Protein , Heme Oxygenase-1/metabolism , In Situ Nick-End Labeling , Membrane Proteins/metabolism , Mice , Mice, Inbred DBA , Mitochondria/metabolism , Nerve Tissue Proteins/metabolism , Real-Time Polymerase Chain Reaction , Receptors, N-Methyl-D-Aspartate/metabolism , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Degeneration/prevention & control , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Superoxide Dismutase/metabolism , Ubiquinone/administration & dosage , Ubiquinone/therapeutic use , Vitamins/administration & dosage , bcl-Associated Death Protein/metabolism
5.
Apoptosis ; 19(4): 603-14, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24337820

ABSTRACT

Coenzyme Q10 (CoQ10) acts by scavenging reactive oxygen species for protecting neuronal cells against oxidative stress in neurodegenerative diseases. We tested whether a diet supplemented with CoQ10 ameliorates oxidative stress and mitochondrial alteration, as well as promotes retinal ganglion cell (RGC) survival in ischemic retina induced by intraocular pressure elevation. A CoQ10 significantly promoted RGC survival at 2 weeks after ischemia. Superoxide dismutase 2 (SOD2) and heme oxygenase-1 (HO-1) expression were significantly increased at 12 h after ischemic injury. In contrast, the CoQ10 significantly prevented the upregulation of SOD2 and HO-1 protein expression in ischemic retina. In addition, the CoQ10 significantly blocked activation of astroglial and microglial cells in ischemic retina. Interestingly, the CoQ10 blocked apoptosis by decreasing caspase-3 protein expression in ischemic retina. Bax and phosphorylated Bad (pBad) protein expression were significantly increased in ischemic retina at 12 h. Interestingly, while CoQ10 significantly decreased Bax protein expression in ischemic retina, CoQ10 showed greater increase of pBad protein expression. Of interest, ischemic injury significantly increased mitochondrial transcription factor A (Tfam) protein expression in the retina at 12 h, however, CoQ10 significantly preserved Tfam protein expression in ischemic retina. Interestingly, there were no differences in mitochondrial DNA content among control- or CoQ10-treated groups. Our findings demonstrate that CoQ10 protects RGCs against oxidative stress by modulating the Bax/Bad-mediated mitochondrial apoptotic pathway as well as prevents mitochondrial alteration by preserving Tfam protein expression in ischemic retina. Our results suggest that CoQ10 may provide neuroprotection against oxidative stress-mediated mitochondrial alterations in ischemic retinal injury.


Subject(s)
Ischemia/drug therapy , Mitochondria/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Retina/metabolism , Ubiquinone/analogs & derivatives , Vitamins/pharmacology , Animals , Apoptosis/drug effects , Body Weight/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diet , Female , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Intraocular Pressure/drug effects , Ischemia/metabolism , Ischemia/pathology , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Neuroprotective Agents/therapeutic use , Phosphorylation , Retina/drug effects , Retina/pathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Ubiquinone/pharmacology , Ubiquinone/therapeutic use , Vitamins/therapeutic use , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , bcl-Associated Death Protein/metabolism
6.
Genes Nutr ; 2(4): 353-8, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18850230

ABSTRACT

The aim of the present study was to investigate the anti-obesity effect of a mixture composed of Garcinia cambogia extract, soypeptide, and L: -carnitine (1.2:0.3:0.02, w/w/w) in rats rendered obese by a high-fat diet (HFD). Sprague-Dawley rats were fed either the high-fat control diet (CD) or the 0.38% mixture-supplemented HFD (CD + M) for 9 weeks. The mixture significantly reduced body weight gain and the accumulation of visceral fat mass in a rat model of HFD-induced obesity. Moreover, the mixture effectively lowered blood and hepatic lipid concentrations and serum glucose, insulin, c-peptide, and leptin levels in rats with HFD-induced obesity. Results from real-time reverse transcription-polymerase chain reaction analyses indicated that the expression levels of leptin, tumor necrosis factor-alpha (TNF-alpha), and sterol regulatory element binding protein 1c (SREBP1c) genes in the epididymal fat tissue of rats fed the CD + M diet were 0.4-, 0.6-, and 0.48-fold, respectively, of those found in the CD rats (P < 0.05), while expression of the uncoupling protein 2 (UCP2) gene in epididymal adipose tissue was 1.25-fold (P < 0.05) of that found in CD rats. In conclusion, a mixture composed of G. cambogia extract, soy peptide, and L: -carnitine attenuated visceral fat accumulation and improved dyslipidemia in a rat model with HFD-induced obesity.

7.
Biosci Biotechnol Biochem ; 72(7): 1772-80, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18603810

ABSTRACT

The aim of present study is to evaluate the effects of Garcinia cambogia on the mRNA levels of the various genes involved in adipogenesis, as well as on body weight gain, visceral fat accumulation, and other biochemical markers of obesity in obesity-prone C57BL/6J mice. Consumption of the Garcinia cambogia extract effectively lowered the body weight gain, visceral fat accumulation, blood and hepatic lipid concentrations, and plasma insulin and leptin levels in a high-fat diet (HFD)-induced obesity mouse model. The Garcinia cambogia extract reversed the HFD-induced changes in the expression pattern of such epididymal adipose tissue genes as adipocyte protein aP2 (aP2), sterol regulatory element-binding factor 1c (SREBP1c), peroxisome proliferator-activated receptor gamma2 (PPARgamma2), and CCAT/enhancer-binding protein alpha (C/EBPalpha). These findings suggest that the Garcinia cambogia extract ameliorated HFD-induced obesity, probably by modulating multiple genes associated with adipogenesis, such as aP2, SREBP1c, PPARgamma2, and C/EBPalpha in the visceral fat tissue of mice.


Subject(s)
Adiposity/drug effects , Dietary Fats/pharmacology , Garcinia cambogia/chemistry , Intra-Abdominal Fat/drug effects , Plant Extracts/pharmacology , Adipogenesis/drug effects , Adipogenesis/genetics , Animals , Dietary Fats/administration & dosage , Gene Expression Profiling , Gene Expression Regulation/drug effects , Insulin/blood , Leptin/blood , Lipids/analysis , Mice , Mice, Inbred C57BL , Plant Extracts/therapeutic use , Weight Gain/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL