Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Publication year range
1.
Biosci Trends ; 17(4): 262-270, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37612125

ABSTRACT

As people age, geriatric syndromes characterized by frailty significantly impact both clinical practice and public health. Aging weakens people's immune functions, leading to chronic low-grade inflammation that ultimately contributes to the development of frailty. Effectively managing geriatric syndromes and frailty can help alleviate the economic burden of an aging population. This review delves into the intricate relationship among aging, infection-induced inflammation, chronic inflammation, and frailty. In addition, it analyzes various approaches and interventions to address frailty, such as smart rehabilitation programs and stem-cell treatments, offering promising solutions in this new era. Given the importance of this topic, further research into the mechanisms of frailty is crucial. Equally essential is the devising of relevant measures to delay its onset and the formulation of comprehensive clinical, research, and public health strategies to enhance the quality of life for elderly individuals.


Subject(s)
Frailty , Aged , Humans , Frailty/therapy , Quality of Life , Syndrome , Aging , Inflammation/therapy
2.
Exp Ther Med ; 9(3): 697-706, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25667616

ABSTRACT

Treatments for leukemia remain unsatisfactory. Conventional chemotherapy agents that aim to kill tumor cells may also damage normal cells and thus result in severe side-effects. Naringenin, a natural polyphenolic compound with antioxidant effects, has been revealed to have significant antitumor effects with low toxicity in preliminary studies. Thus, it is considered as one of the most promising flavonoids in the treatment of leukemia. In the present study, the effects of naringenin on the K562 human leukemia cell line and the underlying mechanisms were explored in vitro. In addition, human peripheral blood polymorphonuclear leukocytes (PMNs) were used as a normal control in order to evaluate the effects of naringenin on normal granulocytes and in the mediation of Adriamycin (ADM)-induced oxidative damage. The results revealed that K562 proliferation was significantly inhibited by naringenin in a time- and concentration-dependent manner; however, minimal cytotoxic effects were observed in PMNs when naringenin was used at concentrations <400 µmol/l. Morphological changes indicative of apoptosis were observed in naringenin-treated K562 cells. Flow cytometric analysis indicated that the K562 cells were arrested in the G0/G1 phase of the cell cycle with a significantly upregulated rate of apoptosis. Furthermore, in the naringenin-treated K562 cells, the labeling index of proliferating cell nuclear antigen was observed to be increased by immunochemical staining, the mRNA and protein expression levels of p21/WAF1 were strongly upregulated in reverse transcription-polymerase chain reaction and western blot analyses, whereas p53 gene expression was not significantly changed. In PMNs to which naringenin (50~80 µmol/l) was added 1 h subsequent to ADM, the cell damage induced by ADM was significantly reduced, coincident with reductions in the levels of reactive oxygen species (ROS) and malondialdehyde (MDA) and increases in the activity of superoxide dismutase and glutathione peroxidase. However, the cytotoxic effect of ADM in K562 cells was not significantly altered by naringenin, and the oxidative stress indices in K562 cells remained stable. In conclusion, the present study revealed the promising value of naringenin in leukemia treatment. Naringenin demonstrated a significant inhibitory effect on the growth of K562 cells but not on normal PMNs. Furthermore, naringenin protected PMNs from ADM-induced oxidative damage at low concentrations. Cell cycle arrest and apoptosis-inducing effects, achieved through p53-independent p21/WAF1 upregulation, are likely to be the mechanism of the antileukemic effects of naringenin, and the protective effect against ADM chemotherapy-induced damage in PMNs may be due to the antioxidant capability of this agent at low concentrations.

3.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 16(4): 790-3, 2008 Aug.
Article in Chinese | MEDLINE | ID: mdl-18718062

ABSTRACT

The objective of this study was to investigate the protection by naringenin against doxorubicin-induced oxidative damage in normal blood cells. Inhibiting effects of naringenin, doxorubicin and naringenin combined with doxorubicind on K562 cells and polymorphonuclear leukocytes were detected with MTT method, the level of reactive oxygen species (ROS) and lipid peroxidation (MDA), the activity of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) were examined with spectrophotometric method in the K562 cells and polymorphonuclear leukocytes. The results indicated that the proliferation of K562 cells was not inhibited by the cytotoxicity of doxorubicin in combination of naringenin with doxorubicin. As compared with the doxorubicin, the addition of naringenin after doxorubicin for 1 hour, the levels of reactive oxygen species (ROS) and lipid peroxidation (MDA) obviously decreased, the activity of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) obviously increased in the polymorphonuclear leukocytes, but these were not changed obviously in K562 cells. It is concluded naringenin can protect against doxorubicin-induced oxidative damage in normal blood cells. The mechanism of naringenin may be elevating activities of antioxidant enzyme and degrading oxidative production level in normal blood cells, and meanswhile decreasing level of oxidative products.


Subject(s)
Antioxidants/pharmacology , Doxorubicin/adverse effects , Erythrocytes/drug effects , Flavanones/pharmacology , Oxidative Stress , Humans , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
4.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 16(2): 286-9, 2008 Apr.
Article in Chinese | MEDLINE | ID: mdl-18426650

ABSTRACT

The objective of this study was to investigate the apoptosis-inducing effect and underlying mechanism of naringenin (NGEN) on K562 cells in vitro. The inhibition of NGEN on K562 cells was evaluated by means of MTT assay so as to observe the cytotoxicity of NGEN; The morphological changes of the cells treated by NGEN were observed by transmission electron microscope; cell apoptosis rate influenced by NGEN was assessed by flow cytometry; the enzyme activity changes of caspase-3 and caspase-8 in the process of NGEN-induced K562 apoptosis were detected by Caspase Colorimetric Assay Kit; immunohistochemistry technique was used to detect FAS, FASL protein expression in K562 cells. The results showed that the growth of cells was inhibited by NGEN in dose-and time-dependent manners (p<0.05); NGEN-induced K562 cells apoptosis and sub-G1 peak was observed; some typically early and final phase changes of cell apoptosis were revealed under transmission electron microscope; the enzyme activity of caspase-3 and caspase-8 and the expression of FAS remarkably increased, meanwhile the expression of FASL was down-regulated (p<0.05). It is concluded that NGEN exerts anti-cancer effect by inducing K562 cell apoptosis, and the regulation of the expression of FAS and FASL. The caspase-3 and caspase-8 co-pathway brings about one of the mechanisms.


Subject(s)
Apoptosis/drug effects , Caspase 3/metabolism , Caspase 8/metabolism , Fas Ligand Protein/metabolism , Flavanones/pharmacology , fas Receptor/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Fas Ligand Protein/genetics , Humans , K562 Cells , fas Receptor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...