Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Kuiyi Xing is active.

Publication


Featured researches published by Kuiyi Xing.


Experimental Eye Research | 2009

Reactive oxygen species (ROS) are essential mediators in epidermal growth factor (EGF)-stimulated corneal epithelial cell proliferation, adhesion, migration, and wound healing.

Yanan Huo; Wen-Ya Qiu; Qing Pan; Yu-Feng Yao; Kuiyi Xing; Marjorie F. Lou

EGF is an essential growth factor needed for epithelial cell proliferation and wound healing of the cornea, but the molecular mechanism is not understood. Although studies have shown that EGF in some non-phagocytic cells induces ROS generation, little is known about the role of ROS in corneal epithelial cells. Therefore, we examined the potential physiological role of ROS in corneal cell proliferation, adhesion and wound healing using rabbit or human corneal epithelial cells, and pig whole cornea organ culture as models. EGF (5 ng/ml)-induced ROS in serum-starved RCE or HCE cells were captured as DCFH fluorescence and detected by confocal microscopy. The elevation of ROS was eradicated when the cells were pretreated with an antioxidant N-acetylcysteine (NAC) or mannitol, or with inhibitor to NADPH oxidase (DPI), or to lipoxygenase (NDGA). EGF-induced ROS generation correlated with cell growth and activation of Akt and MAPK signaling pathways, while NAC eliminated all these effects. EGF-stimulated cell adhesion or migration in cell culture was greatly suppressed in the presence of NAC while EGF-facilitated epithelial cell wound healing in corneal organ culture was also blocked by NAC. This is the first demonstration of a novel ROS physiological function in corneal wound healing.


Biochimica et Biophysica Acta | 2010

Glutaredoxin 2 prevents H2O2-induced cell apoptosis by protecting complex I activity in the mitochondria☆

Hongli Wu; Kuiyi Xing; Marjorie F. Lou

Glutaredoxin 2 (Grx2) belongs to the oxidoreductase family and is an isozyme of glutaredoxin 1 (Grx1) present in the mitochondria, however its function is not well understood. The purpose of this study is to evaluate the potential anti-apoptotic function of Grx2 by examining its ability to protect complex I in the mitochondrial electron transport system using human lens epithelial cells as a model. We found that cells treated with 200muM hydrogen peroxide (H(2)O(2)) for 24h exhibited decreased viability and became apoptotic with corresponding Bax up-regulation, Bcl-2 down-regulation, caspase 3 activation and mitochondrial cytochrome c leakage. Grx2 over-expression (OE) could protect cells against H(2)O(2)-induced damage while Grx2 knockdown (KD) showed the opposite effect. Under the same conditions, H(2)O(2) treatment caused 50% inactivation of complex I activity in control cells (vector only), 75% in Grx2 KD cells but only 20% in Grx2 OE cells. Furthermore, the inactivated complex I in the H(2)O(2)-treated cells could be protected mostly by importing the purified nascent Grx2 protein, but not the Grx2 protein mutated at the active site with C70S, or C73S, or with C70S plus C73S. Immunoprecipitation study also revealed that Grx2 co-precipitated with complex I, but not complex II, in the mitochondrial lysate. Thus, the mechanism of Grx2 protection against H(2)O(2)-induced apoptosis is likely associated with its ability to preserve complex I.


The FASEB Journal | 2003

The possible physiological function of thioltransferase in cells

Kuiyi Xing; Marjorie F. Lou

We sought to study the possible physiological function of thioltransferase (TTase) in combating oxidative damage in the lens epithelial cells. The cells transfected with either TTase‐containing plasmid or vector only were compared for their resistance to oxidative stress in the presence of a bolus of H2O2 (0.1 mM) for 3 h. Cells depleted of TTase activity upon cadmium treatment were also examined for the resistance to oxidative stress under the same conditions. TTase activity assay, Western blot, and Northern blot analyses confirmed that hTTase gene was successfully transfected into the HLE B3 cells and was overexpressed. The TTase‐transfected cells detoxified H2O2 as efficiently as the control cells but displayed a faster and more complete recovery of oxidatively inactivated glyceraldehyde 3‐phosphate dehydrogenase (GAPDH) and glutathione peroxidase (GPx) activities and suppressed protein thiolation (PSSG formation). With TTase activity being inhibited by cadmium, the spontaneous reactivation of GAPDH under bolus H2O2 treatment was not accomplished in cadmium‐pretreated cells. These data indicate a new physiological function of TTase, which involves in the reactivation of the oxidatively inactivated enzymes through dethiolation; thus this redox‐regulating enzyme can protect the human lens epithelial cells and maybe other cell types by preventing them from permanent oxidative damage.


Investigative Ophthalmology & Visual Science | 2010

Effect of age on the thioltransferase (glutaredoxin) and thioredoxin systems in the human lens.

Kuiyi Xing; Marjorie F. Lou

PURPOSE To investigate the effect of age on the key oxidation repair enzymes of the thioltransferase (TTase) and thioredoxin (TRx) systems in the human lens. METHODS Twenty-three normal human lenses (donor ages, 19-77 years) were grouped into second, third, fifth, sixth, and seventh decades and analyzed for TTase, TRx, glutathione reductase (GR), thioredoxin reductase (TR), and glyceraldehyde-3-phosphate dehydrogenase (G3PD) activities, as well as the glutathione (GSH) pool. Additionally, 19 contralateral lenses of the donor eyes were each divided into cortex and nucleus for enzyme distribution studies. RESULTS All the enzymes showed similar activity in the cortex and nucleus, regardless of age, but were inactivated to various extents in the older lenses. In the TTase system, both TTase and GR showed activity loss over the five decades, with 70% remaining in the seventh decade, whereas the GSH pool was depleted extensively, with only 35% left in the older lenses. In the TRx system, TRx activity was not affected as much as TR for which only 70% of the activity was found in the seventh decade compared with the second to third decades. Overall, G3PD was more sensitive to age because only 50% activity remained after the sixth decade. CONCLUSIONS With increasing age there is a gradual activity loss in both the TTase and the TRx systems and a lowered GSH pool. These alterations, compounded with the age-related loss in G3PD activity, may lead to redox and energy imbalance, likely contributing to a higher risk to cataract formation in the aging population.


Experimental Eye Research | 2012

Osmotic stress, not aldose reductase activity, directly induces growth factors and MAPK signaling changes during sugar cataract formation.

Peng Zhang; Kuiyi Xing; James Randazzo; Karen Blessing; Marjorie F. Lou; Peter F. Kador

In sugar cataract formation in rats, aldose reductase (AR) activity is not only linked to lenticular sorbitol (diabetic) or galactitol (galactosemic) formation but also to signal transduction changes, cytotoxic signals and activation of apoptosis. Using both in vitro and in vivo techniques, the interrelationship between AR activity, polyol (sorbitol and galactitol) formation, osmotic stress, growth factor induction, and cell signaling changes have been investigated. For in vitro studies, lenses from Sprague Dawley rats were cultured for up to 48 h in TC-199-bicarbonate media containing either 30 mM fructose (control), or 30 mM glucose or galactose with/without the aldose reductase inhibitors AL1576 or tolrestat, the sorbitol dehydrogenase inhibitor (SDI) CP-470,711, or 15 mM mannitol (osmotic-compensated media). For in vivo studies, lenses were obtained from streptozotocin-induced diabetic Sprague Dawley rats fed diet with/without the ARIs AL1576 or tolrestat for 10 weeks. As expected, lenses cultured in high glucose/galactose media or from untreated diabetic rats all showed a decrease in the GSH pool that was lessened by ARI treatment. Lenses either from diabetic rats or from glucose/galactose culture conditions showed increased expression of basic-FGF, TGF-β, and increased signaling through P-Akt, P-ERK1/2 and P-SAPK/JNK which were also normalized by ARIs to the expression levels observed in non-diabetic controls. Culturing rat lenses in osmotically compensated media containing 30 mM glucose or galactose did not lead to increased growth factor expression or altered signaling. These studies indicate that it is the biophysical response of the lens to osmotic stress that results in an increased intralenticular production of basic-FGF and TGF-β and the altered cytotoxic signaling that is observed during sugar cataract formation.


Current Eye Research | 2007

Revival of Glutathione Reductase in Human Cataractous and Clear Lens Extracts by Thioredoxin and Thioredoxin Reductase, in Conjunction with α-Crystallin or Thioltransferase

Hong Yan; John J. Harding; Kuiyi Xing; Marjorie F. Lou

Glutathione reductase (GR) plays a key role in maintaining thiol groups in the lens, and its activity decreases with aging and cataract formation. Mammalian thioredoxin (Trx) and thioredoxin reductase (TrxR), or the Trx/TrxR system, participates in the repair of oxidatively damaged lens proteins and enzymes. α -Crystallin, a molecular chaperone, prevents the aggregation of partially denatured proteins under various stress conditions. Thioltransferase (TTase, or glutaredoxin) can maintain the homeostasis of lens protein thiols thus protecting against oxidative stress. We investigated whether the Trx/TrxR system can revive GR activity in both the cortex and nucleus of human cataract and clear aged lenses and whether α -crystallin and TTase can help this effect. The GR activity in the cortex and nucleus of the cataractous lenses was significantly lower than that of the aged clear lenses. The highest activity in the cortex was observed in the clear aged lenses. The combination of Trx and TrxR revived the activity of GR from both the cortex and nucleus of aged clear lenses. However, in cataract lenses (grade II and grade IV), there was a statistically significant recovery of GR activity in the cortex, but not in the nucleus. No recovery was observed when Trx or TrxR were used separately. α -Crystallin successfully revived GR activity in the cortex of cataract grade II lenses, but not in the nucleus. The combination of α -crystallin and Trx/TrxR gave a further increase of activity. TTase alone revived some of the GR activity but together with the Trx/TrxR system gave no statistically significant enhancement of GR activity. These results indicate that both disulfide bond formation and protein unfolding are responsible for GR inactivation.


Free Radical Biology and Medicine | 2013

Overexpression of thioredoxin-binding protein 2 increases oxidation sensitivity and apoptosis in human lens epithelial cells.

Yibo Yu; Kuiyi Xing; Rilwan Badamas; Charles A. Kuszynski; Hongli Wu; Marjorie F. Lou

Thioredoxin (Trx) is an important redox regulator with cytosolic Trx1 and mitochondrial Trx2 isozymes. Trx has multiple physiological functions in cells and its bioavailability is negatively controlled through active-site binding to a specific thioredoxin-binding protein (TBP-2). This paper describes the delicate balance between TBP-2 and Trx and the effect of overexpression of TBP-2 in human lens epithelial cells. Cells overexpressing TBP-2 (TBP-2 OE) showed a sevenfold increase in TBP-2 and a nearly 40% suppression of Trx activity but no change in Trx expression. The TBP-2 OE cells grew slower and their population decreased to 30% by day 7. Cell cycle analysis showed that TBP-2 OE cells arrested at the G2/M stage and that they displayed low expression of the cell cycle elements P-cdc2(Y15), cdc2, cdc25A, and cdc25C. Furthermore, TBP-2 OE cells were more sensitive to oxidation. Under H2O2 (200μM, 24h) treatment, these cells lost 80% viability and became highly apoptotic. Brief oxidative stress (200μM, 30min) to TBP-2 OE cells disrupted the Trx antiapoptotic function by dissociating the cytosolic and mitochondrial Trx-ASK binding complexes. The same H2O2-treated cells also showed activated ASK (P-ASK), increased Bax, lowered Bcl-2, cytochrome c release, and elevated caspase 3/7 activity. We conclude from these studies that high cellular levels of TBP-2 can potentially suppress Trx bioavailability and increase oxidation sensitivity. Overexpression of TBP-2 also causes slow growth by mitotic arrest and apoptosis by activating the ASK death pathway.


Experimental Eye Research | 2004

Platelet derived growth factor (PDGF)-induced reactive oxygen species in the lens epithelial cells: the redox signaling.

Kate Chao Wei Chen; You Zhou; Kuiyi Xing; Kostantyn Krysan; Marjorie F. Lou


Experimental Eye Research | 2002

Effect of H2O2on Human Lens Epithelial Cells and the Possible Mechanism for Oxidative Damage Repair by Thioltransferase

Kuiyi Xing; Marjorie F. Lou


Investigative Ophthalmology & Visual Science | 2001

Human Lens Thioltransferase: Cloning, Purification, and Function

Fengyu Qiao; Kuiyi Xing; Aimin Liu; Nancy Ehlers; Nalini Raghavachari; Marjorie F. Lou

Collaboration


Dive into the Kuiyi Xing's collaboration.

Top Co-Authors

Avatar

Marjorie F. Lou

University of Nebraska–Lincoln

View shared research outputs
Top Co-Authors

Avatar

Hongli Wu

University of Nebraska–Lincoln

View shared research outputs
Top Co-Authors

Avatar

Fengyu Qiao

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

James Randazzo

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Min Wei

University of Nebraska–Lincoln

View shared research outputs
Top Co-Authors

Avatar

Nalini Raghavachari

University of Nebraska–Lincoln

View shared research outputs
Top Co-Authors

Avatar

Ye-Shih Ho

Wayne State University

View shared research outputs
Top Co-Authors

Avatar

Yin Wang

University of Nebraska–Lincoln

View shared research outputs
Top Co-Authors

Avatar

Aimin Liu

University of Nebraska–Lincoln

View shared research outputs
Top Co-Authors

Avatar

Ashraf S. Raza

University of Nebraska–Lincoln

View shared research outputs
Researchain Logo
Decentralizing Knowledge