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










Publication year range
1.
Sci Rep ; 11(1): 12457, 2021 06 14.
Article in English | MEDLINE | ID: mdl-34127701

ABSTRACT

Burn wound progression is an important determinant of patient morbidity and mortality after injury. In this study, we used the brass comb contact burn to determine burn wound vertical injury progression with a focus on blood vessel occlusion and endothelial cell death. Class A 3-month-old Yorkshire pigs received a brass comb contact burn. Burn wounds were sampled at 0, 30 min, 1, 2, 4, and 24 h. Hematoxylin Phloxin Saffron staining and vimentin immunostaining were performed to determine the depth of blood vessel occlusion and endothelial cell death, respectively. The depth of blood vessel occlusion increased by 30 min (p < 0.005) and peaked by 1 to 4 h (p > 0.05). The depth of endothelial cell death risen to a plateau at 30 min (p < 0.005) to 2 h and then peaked at 24 h (p < 0.03). We observed a progression of blood vessel occlusion and vascular endothelial cell death from the middle of the dermis to the hypodermis within 2 h to 4 h after the initial injury, namely a progression from a second-degree (partial thickness) to third-degree (full thickness) burn. These data suggest that therapeutic interventions during this time window may provide a better outcome by reducing or preventing vertical progression of blood vascular occlusion or endothelial cell death.


Subject(s)
Burns/diagnosis , Endothelium, Vascular/pathology , Skin/blood supply , Vascular Patency , Animals , Burns/pathology , Burns/therapy , Disease Models, Animal , Disease Progression , Endothelial Cells/pathology , Hot Temperature/adverse effects , Humans , Injury Severity Score , Skin/pathology , Sus scrofa , Time-to-Treatment , Wound Healing
2.
Redox Biol ; 45: 102034, 2021 09.
Article in English | MEDLINE | ID: mdl-34139550

ABSTRACT

Oxidative stress and inflammation may mediate cellular damage and tissue destruction as the burn wound continues to progress after the abatement of the initial insult. Since iron and calcium ions play key roles in oxidative stress, this study tested whether topical application of a metal chelator proprietary lotion (Livionex Formulation (LF) lotion), that contains disodium EDTA as a metal chelator and methyl sulfonyl methane (MSM) as a permeability enhancer, would prevent progression or reduce burn wound severity in a porcine model. We have reported earlier that in a rat burn model, LF lotion reduces thermal injury progression. Here, we used the porcine brass comb burn model that closely mimics the human condition for contact burns and applied LF lotion every 8 h starting 15 min after the injury. We found that LF lotion reduces the depth of cell death as assessed by TUNEL staining and blood vessel blockage in the treated burn sites and interspaces. The protein expression of pro-inflammatory markers IL-6, TNF-a, and TNFα Converting Enzyme (TACE), and lipid aldehyde production (protein-HNE) was reduced with LF treatment. LF lotion reversed the burn-induced decrease in the aldehyde dehydrogenase (ALDH-1) expression in the burn sites and interspaces. These data show that a topically applied EDTA-containing lotion protects both vertical and horizontal burn progression when applied after thermal injury. Curbing burn wound conversion and halting the progression of second partial burn to third-degree full-thickness burn remains challenging when it comes to burn treatment strategies during the acute phase. Burn wound conversion can be reduced with targeted treatments to attenuate the oxidative and inflammatory response in the immediate aftermath of the injury. Our studies suggest that LF lotion could be such a targeted treatment.


Subject(s)
Burns , Animals , Burns/drug therapy , Chelating Agents , Copper , Disease Models, Animal , Inflammation/drug therapy , Oxidative Stress , Rats , Swine , Zinc
3.
Burns Trauma ; 8: tkaa024, 2020.
Article in English | MEDLINE | ID: mdl-33033727

ABSTRACT

BACKGROUND: One of the most pervasive complications of burn injury is wound progression, characterized by continuous tissue destruction in untreated wounds, which leads to wound infection, inflammation, oxidative stress and excessive scar formation. We determined whether additional tissue destruction could be attenuated with Livionex formulation (LF) lotion, which contains a metal-chelating agent and reduces inflammation in burn wounds. METHODS: We subjected male Sprague Dawley rats to a 2% total body surface area (TBSA) burn using a brass comb model and topically applied LF lotion (containing ethylenediaminetetraacetic acid and methyl sulfonyl methane) to the affected area every 8 hours over 3 days. Inflammatory cytokine levels, cell apoptosis and wound healing were compared in LF lotion-treated and untreated rats. Statistical analysis was performed using a one-way analysis of variance in conjunction with Tukey's post-hoc test. RESULTS: Serum inflammatory cytokines were not detectable after 3 days, suggesting that small burn wounds induce only an immediate, localized inflammatory response. Microscopy revealed that LF lotion improved burn site pathology. Deoxynucleotidyl transferase biotin-d-UTP nick-end labeling staining showed reduced cell death in the LF-treated samples. LF lotion prevented the spread of tissue damage, as seen by increased amounts of Ki-67-positive nuclei in the adjacent epidermis and hair follicles. Tumor necrosis factor-alpha, interleukin-6 and inducible nitric oxide synthase levels in LF-treated skin sections from burned rats were comparable to the levels observed in unburned control sections, indicating that LF lotion reduces inflammation in and around the burn site. CONCLUSIONS: These results establish LF lotion as a therapeutic agent for reducing inflammatory stress, cell death and tissue destruction when applied immediately after a burn injury. Further studies of LF lotion on large TBSA burns will determine its efficacy as an emergency treatment for reducing long-term morbidity and scarring.

4.
Invest Ophthalmol Vis Sci ; 59(1): 31-38, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29302691

ABSTRACT

Purpose: Metal ions play a key role in exacerbating toxicity associated with oxidative stress and inflammation. This study examines the effects of a formulation containing the metal chelator ethylenediaminetetraacetic acid (EDTA) and permeability enhancer methyl sulfonyl methane (MSM) on the early course of inflammation in endotoxin-induced uveitis (EIU). The proprietary MSM/EDTA formulation of Livionex, Inc., which was used for this study, is covered by several patents and pending patent applications. Methods: EIU was induced by using subcutaneous injection of lipopolysaccharide (LPS) into the thighs of Lewis rats. Treatment consisted of topical application to the eyes of either PBS or eye drops designated as ME that contain EDTA and MSM. Clinical signs of uveitis were monitored at 6 and 24 hours postinjection. Oxidative and inflammatory markers were evaluated by ELISA or immunohistochemistry. Results: Rats treated with ME showed fewer clinical signs of uveitis including reduced miosis, fibrinous exudates, and dilated blood vessels. The aqueous humor of treated rats contained fewer leukocytes, lower protein levels, and less PGE2. Formation of protein adducts with the lipid peroxidation end-product, 4-hydroxynonenal, expression of NF-κB, TNF-α, and MMP-9 were all reduced in rats treated with ME. Conclusions: Our results indicate that ME eye drops downregulate the ocular inflammatory response in LPS treated rats, suggesting that induction of EIU involves metal ions and chelation therapy with ME is a potential treatment for uveitis.


Subject(s)
Aqueous Humor/metabolism , Cytokines/biosynthesis , Edetic Acid/therapeutic use , Oxidative Stress , Uveitis/drug therapy , Animals , Aqueous Humor/drug effects , Calcium Chelating Agents/therapeutic use , Cytokines/drug effects , Disease Models, Animal , Male , Rats , Rats, Inbred Lew , Uveitis/diagnosis , Uveitis/metabolism
6.
Aging (Albany NY) ; 7(9): 718-33, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26423835

ABSTRACT

Functional competence and self-renewal of mammalian skeletal muscle myofibers and progenitor cells declines with age. Progression of the muscle aging phenotype involves the decline of juvenile protective factorsi.e., proteins whose beneficial functions translate directly to the quality of life, and self-renewal of progenitor cells. These characteristics occur simultaneously with the age-associated increase of p38α stress response signaling. This suggests that the maintenance of low levels of p38α activity of juvenile tissues may delay or attenuate aging. We used the dominant negative haploinsufficient p38α mouse (DN-p38α(AF/+)) to demonstrate that in vivo attenuation of p38α activity in the gastrocnemius of the aged mutant delays age-associated processes that include: a) the decline of the juvenile protective factors, BubR1, aldehyde dehydrogenase 1A (ALDH1A1), and aldehyde dehydrogenase 2 (ALDH2); b) attenuated expression of p16(Ink4a) and p19(Arf) tumor suppressor genes of the Cdkn2a locus; c) decreased levels of hydroxynonenal protein adducts, expression of COX2 and iNOS; d) decline of the senescent progenitor cell pool level and d) the loss of gastrocnemius muscle mass. We propose that elevated P-p38α activity promotes skeletal muscle aging and that the homeostasis of p38α impacts the maintenance of a beneficial healthspan.


Subject(s)
Aging/genetics , Aging/pathology , Mitogen-Activated Protein Kinase 14/genetics , Muscle Fibers, Skeletal/pathology , Stem Cells/pathology , Stress, Physiological , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase 1 Family , Aldehyde Dehydrogenase, Mitochondrial , Animals , Cell Cycle Proteins/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclooxygenase 2/genetics , Female , Male , Mice , Mice, Knockout , Muscle, Skeletal/pathology , Nitric Oxide Synthase Type II/genetics , Protein Serine-Threonine Kinases/genetics , Retinal Dehydrogenase , Signal Transduction
7.
Burns ; 41(8): 1775-1787, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26392023

ABSTRACT

UNLABELLED: Oxidative stress may be involved in the cellular damage and tissue destruction as burn wounds continues to progress after abatement of the initial insult. Since iron and calcium ions play key roles in oxidative stress, this study tested whether topical application of Livionex formulation (LF) lotion, that contains disodium EDTA as a metal chelator and methyl sulfonyl methane (MSM) as a permeability enhancer, would prevent or reduce burns. METHODS: We used an established brass comb burn model with some modifications. Topical application of LF lotion was started 5 min post-burn, and repeated every 8 h for 3 consecutive days. Rats were euthanized and skin harvested for histochemistry and immunohistochemistry. Formation of protein adducts of 4-hydroxynonenal (HNE), malonadialdehyde (MDA) and acrolein (ACR) and expression of aldehyde dehydrogenase (ALDH) isozymes, ALDH1 and ALDH2 were assessed. RESULTS: LF lotion-treated burn sites and interspaces showed mild morphological improvement compared to untreated burn sites. Furthermore, the lotion significantly decreased the immunostaining of lipid aldehyde-protein adducts including protein -HNE, -MDA and -ACR adducts, and restored the expression of aldehyde dehydrogenase isozymes in the unburned interspaces. CONCLUSION: This data, for the first time, demonstrates that a topically applied EDTA-containing lotion protects burns progression with a concomitant decrease in the accumulation of reactive lipid aldehydes and protection of aldehyde dehydrogenase isozymes. Present studies are suggestive of therapeutic intervention of burns by this novel lotion.


Subject(s)
Burns , Chelating Agents/pharmacology , Dimethyl Sulfoxide/pharmacology , Edetic Acid/pharmacology , Oxidative Stress/drug effects , Skin/drug effects , Sulfones/pharmacology , Acrolein/metabolism , Administration, Cutaneous , Aldehyde Dehydrogenase/drug effects , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family , Aldehyde Dehydrogenase, Mitochondrial , Aldehydes/metabolism , Animals , Copper , Disease Models, Animal , Immunohistochemistry , Malondialdehyde/metabolism , Mitochondrial Proteins/drug effects , Mitochondrial Proteins/metabolism , Permeability/drug effects , Rats , Retinal Dehydrogenase/drug effects , Retinal Dehydrogenase/metabolism , Skin/metabolism , Skin/pathology , Trauma Severity Indices , Zinc
8.
Curr Med Chem ; 21(2): 230-7, 2014.
Article in English | MEDLINE | ID: mdl-23848536

ABSTRACT

Metastable aldehydes produced by lipid peroxidation act as 'toxic second messengers' that extend the injurious potential of free radicals. 4-hydroxy 2-nonenal (HNE), a highly toxic and most abundant stable end product of lipid peroxidation, has been implicated in the tissue damage, dysfunction, injury associated with aging and other pathological states such as cancer, Alzheimer, diabetes, cardiovascular and inflammatory complications. Further, HNE has been considered as a oxidative stress marker and it act as a secondary signaling molecule to regulates a number of cell signaling pathways. Biological activity of HNE depends on its intracellular concentration, which can differentially modulate cell death, growth and differentiation. Therefore, the mechanisms responsible for maintaining the intracellular levels of HNE are most important, not only in the defense against oxidative stress but also in the pathophysiology of a number of disease processes. In this review, we discussed the significance of HNE in mediating various disease processes and how regulation of its metabolism could be therapeutically effective.


Subject(s)
Aldehydes/metabolism , Disease Progression , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Humans , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Lipid Peroxidation , Molecular Structure , Neoplasms/metabolism , Neoplasms/pathology
9.
Invest Ophthalmol Vis Sci ; 53(7): 3431-40, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22562515

ABSTRACT

PURPOSE: To investigate the therapeutic effects of metformin, a commonly used antidiabetic drug, in preventing endotoxin-induced uveitis (EIU) in rats. METHODS: EIU in Lewis rats was developed by subcutaneous injection of lipopolysaccharide (LPS; 150 µg). Metformin (300 mg/kg body weight, intraperitoneally) or its carrier was injected either 12 hours before or 2 hours after LPS induction. Three and 24 hours after EIU, eyes were enucleated and aqueous humor (AqH) was collected. The MILLIPLEX-MAG Rat cytokine-chemokine magnetic bead array was used to determine inflammatory cytokines. The expression of Cox-2, phosphorylation of AMPK, and NF-κB (p65) were determined immunohistochemically. Primary human nonpigmented ciliary epithelial cells (HNPECs) were used to determine the in vitro efficacy of metformin. RESULTS: Compared with controls, the EIU rat AqH had significantly increased number of infiltrating cells and increased levels of various cytokines and chemokines (TNF-α, MCP-1, IL-1ß, MIP-1α, IL-6, Leptin, and IL-18) and metformin significantly prevented the increase. Metformin also prevented the expression of Cox-2 and phosphorylation of p65, and increased the activation of AMPK in the ciliary bodies and retinal tissues. Moreover, metformin prevented the expression of Cox-2, iNOS, and activation of NF-kB in the HNPECs and decreased the levels of NO and PGE2 in cell culture media. CONCLUSIONS: Our results for the first time demonstrate a novel role of the antidiabetic drug, metformin, in suppressing uveitis in rats and suggest that this drug could be developed to prevent uveitis complications.


Subject(s)
Disease Models, Animal , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Uveitis/prevention & control , AMP-Activated Protein Kinase Kinases , Animals , Aqueous Humor/metabolism , Blotting, Western , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Injections, Intraperitoneal , Lipopolysaccharides , Male , Phosphorylation , Protein Kinases/metabolism , Rats , Rats, Inbred Lew , Transcription Factor RelA/metabolism , Uveitis/chemically induced , Uveitis/metabolism
10.
Invest Ophthalmol Vis Sci ; 52(8): 5144-52, 2011 Jul 11.
Article in English | MEDLINE | ID: mdl-21551413

ABSTRACT

PURPOSE: Recent studies indicate that ethyl pyruvate (EP) exerts anti-inflammatory properties; however, the effect of EP on ocular inflammation is not known. The efficacy of EP in endotoxin-induced uveitis (EIU) in rats was investigated. METHODS: EIU in Lewis rats was developed by the subcutaneous injection of lipopolysaccharide (LPS; 150 µg). EP (30 mg/kg body weight) or its carrier was injected intraperitoneally 1 hour before or 2 hours after lipopolysaccharide injection. Animals were killed after 3 and 24 hours followed by enucleation of eyes and collection of the aqueous humor (AqH). The number of infiltrating cells and levels of proteins in the AqH were determined. The rat cytokine/chemokine multiplex method was used to determine level of cytokines and chemokines in the AqH. TNF-α and phospho-nuclear factor kappa B (NF-κB) expression in ocular tissues were determined immunohistochemically. Human primary nonpigmented ciliary epithelial cells (HNPECs) were used to determine the in vitro efficacy of EP on lipopolysaccharide-induced inflammatory response. RESULTS: Compared to controls, AqH from the EIU rat eyes had a significantly higher number of infiltrating cells, total protein, and inflammatory cytokines/chemokines, and the treatment of EP prevented EIU-induced increases. In addition, EP also prevented the expression of TNF-α and activation of NF-κB in the ciliary bodies and retina of the eye. Moreover, in HNPECs, EP inhibited lipopolysaccharide-induced activation of NF-κB and expression of Cox-2, inducible nitric oxide synthase, and TNF-α. CONCLUSIONS: Our results indicate that EP prevents ocular inflammation in EIU, suggesting that the supplementation of EP could be a novel approach for the treatment of ocular inflammation, specifically uveitis.


Subject(s)
Pyruvates/therapeutic use , Uveitis/prevention & control , Animals , Aqueous Humor/metabolism , Blotting, Western , Cell Survival , Cells, Cultured , Ciliary Body/cytology , Cyclooxygenase 2/metabolism , Disease Models, Animal , Endotoxins/toxicity , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Lipopolysaccharides/toxicity , Male , Microscopy, Fluorescence , NF-kappa B/metabolism , Rats , Rats, Inbred Lew , Reactive Oxygen Species/metabolism , Treatment Outcome , Tumor Necrosis Factor-alpha/metabolism , Uveitis/chemically induced , Uveitis/metabolism
11.
Chem Biol Interact ; 191(1-3): 330-8, 2011 May 30.
Article in English | MEDLINE | ID: mdl-21354119

ABSTRACT

Oxidative stress-induced inflammation is a major contributor to several disease conditions including sepsis, carcinogenesis and metastasis, diabetic complications, allergic asthma, uveitis and after cataract surgery posterior capsular opacification. Since reactive oxygen species (ROS)-mediated activation of redox-sensitive transcription factors and subsequent expression of inflammatory cytokines, chemokines and growth factors are characteristics of inflammatory disorders, we envisioned that by blocking the molecular signals of ROS that activate redox-sensitive transcription factors, various inflammatory diseases could be ameliorated. We have indeed demonstrated that ROS-induced lipid peroxidation-derived lipid aldehydes such as 4-hydroxy-trans-2-nonenal (HNE) and their glutathione-conjugates (e.g. GS-HNE) are efficiently reduced by aldose reductase to corresponding alcohols which mediate the inflammatory signals. Our results showed that inhibition of aldose reductase (AKR1B1) significantly prevented the inflammatory signals induced by cytokines, growth factors, endotoxins, high glucose, allergens and auto-immune reactions in cellular as well as animal models. We have demonstrated that AKR1B1 inhibitor, fidarestat, significantly prevents tumor necrosis factor-alpha (TNF-α)-, growth factors-, lipopolysachharide (LPS)-, and environmental allergens-induced inflammatory signals that cause various inflammatory diseases. In animal models of inflammatory diseases such as diabetes, cardiovascular, uveitis, asthma, and cancer (colon, breast, prostate and lung) and metastasis, inhibition of AKR1B1 significantly ameliorated the disease. Our results from various cellular and animal models representing a number of inflammatory conditions suggest that ROS-induced inflammatory response could be reduced by inhibition of AKR1B1, thereby decreasing the progression of the disease and if the therapy is initiated early, the disease could be eliminated. Since fidarestat has already undergone phase III clinical trial for diabetic neuropathy and found to be safe, though clinically not very effective, our results indicate that it can be developed for the therapy of a number of inflammation-related diseases. Our results thus offer a novel therapeutic approach to treat a wide array of inflammatory diseases.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Inflammation/drug therapy , Inflammation/metabolism , Oxidative Stress/drug effects , Aldehyde Reductase/metabolism , Animals , Antioxidants/metabolism , Enzyme Inhibitors/therapeutic use , Humans , Inflammation/pathology , Inflammation/physiopathology
12.
J Toxicol Environ Health A ; 74(6): 380-91, 2011.
Article in English | MEDLINE | ID: mdl-21271438

ABSTRACT

Oxidative stress plays a critical role in cataractogenesis, the leading cause of blindness worldwide. Since transition metals generate reactive oxygen species (ROS) formation, metal chelation therapy has been proposed for treatment of cataracts. However, the effectiveness of most chelators is limited by low tissue penetrability. This study is the first to demonstrate that the topically applied divalent metal chelator ethylenediamine tetraacetic acid (EDTA) combined with the carrier and permeability enhancer methyl sulfonyl methane (MSM) ameliorates both oxidation-induced lens opacification and the associated toxic accumulation of protein-4-hydroxynonenal (HNE) adducts. Both in vitro (rat lens culture) and in vivo (diabetic rats), EDTA-MSM (1) significantly reduced lens opacification by about 40-50%, (2) significantly diminished lens epithelial cell proliferation and fiber cell swelling in early stages of cataract formation in vivo, and (3) notably decreased the levels of protein-HNE adducts. These findings have important implications specifically for the treatment of cataract and generally for other diseases in which oxidative stress plays a key pathogenic role.


Subject(s)
Cataract/drug therapy , Chelating Agents/therapeutic use , Chelation Therapy/methods , Diabetes Complications/drug therapy , Lens, Crystalline/drug effects , Metals/metabolism , Administration, Topical , Aldehydes/toxicity , Animals , Cataract/metabolism , Cataract/pathology , Cell Proliferation/drug effects , Chelating Agents/administration & dosage , Chelating Agents/metabolism , Diabetes Complications/chemically induced , Diabetes Complications/pathology , Dimethyl Sulfoxide/administration & dosage , Dimethyl Sulfoxide/metabolism , Dimethyl Sulfoxide/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Edetic Acid/administration & dosage , Edetic Acid/metabolism , Edetic Acid/therapeutic use , Epithelial Cells/metabolism , Epithelial Cells/pathology , Lens, Crystalline/ultrastructure , Male , Oxidation-Reduction , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Sulfones/administration & dosage , Sulfones/metabolism , Sulfones/therapeutic use
13.
Zhonghua Yan Ke Za Zhi ; 45(3): 248-53, 2009 Mar.
Article in Chinese | MEDLINE | ID: mdl-19575921

ABSTRACT

OBJECTIVE: To study the metabolism of 4-hydroxynonenal (HNE), one of lipid derived aldehydes (LDAs), in diabetic rat lens and its role in diabetic cataract formation. METHODS: Experimental research. A factor design was used to set up the experiment statistically upon two factors: diabetic and normal control as treatment factors; day 30, 45 and 70 as the time factors. Normal and diabetic rats' lenses were incubated with HNE for 2 hours. HNE metabolites in the culture media were studied by high performance liquid chromatography (HPLC). Aldehyde dehydrogenase (ALDH) activity in normal and diabetic rat lens (30, 45 and 70 d after inducing of cataract) was detected by a spectrophotometer, ALDH protein and HNE-protein were detected by Western Blot. All data were analyzed by the Bonferroni test using SAS 8.0 software. RESULTS: The major pathway for HNE metabolism in normal lens was conjugation with glutathione (GSH) to form GS-HNE (45%), followed by HNE's oxidation to 4-hydroxy-2-nonenoic acid (HNA) by ALDH, which accounted for approximately 9.1% of HNE. The conjugation of HNE with GSH in diabetic lens was decreased approximately 64% at day 30 compared with the controls (F = 49.59, P < 0.001). The pathway of HNE oxidation by ALDH in the diabetic lens was enhanced approximately 1.7 times at day 70 compared to day 30 (F = 11.51, P = 0.0442). A higher ALDH activity, greater amount of ALDH protein, and less amount of HNE-protein adduct were presented in diabetic rat lens. CONCLUSIONS: The pathway of conjugation of HNE with GSH is inhibited in diabetic lens which may play a role in the formation of diabetic cataract. The oxidation of HNE by ALDH is a compensation process for protecting the lens against diabetic damage.


Subject(s)
Aldehydes/metabolism , Cataract/metabolism , Diabetes Complications/metabolism , Aldehyde Dehydrogenase/metabolism , Animals , Oxidation-Reduction , Rats , Rats, Sprague-Dawley
14.
Drug Deliv ; 16(5): 243-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19538004

ABSTRACT

Pharmacologic chelators do not effectively penetrate cell membranes and blood-brain barrier. This study assesses methylsulfonylmethane (MSM) as a permeability enhancer and an excipient to facilitate EDTA transport across biologic membranes, and to make possible localized, regional chelation. Topical application of MSM with C(14)EDTA onto the rat cornea led to uptake of the C(14)EDTA in all tested ocular tissues. Without MSM, EDTA did not penetrate the eye. The ability of MSM to deliver EDTA into an eye provides an opportunity for regional chelation therapy. Additionally, these studies suggest that MSM could also be an adjuvant for delivering ciprofloxacin and other chemical compounds to specific, local tissue sites.


Subject(s)
Blood-Brain Barrier/drug effects , Chelating Agents/administration & dosage , Chelation Therapy , Dimethyl Sulfoxide/administration & dosage , Edetic Acid/administration & dosage , Eye/drug effects , Sulfones/administration & dosage , Animals , Chelating Agents/pharmacology , Cornea/drug effects , Cornea/metabolism , Drug Synergism , Edetic Acid/chemistry , Eye/metabolism , Male , Radioisotopes , Rats , Rats, Sprague-Dawley
15.
J Toxicol Environ Health A ; 72(9): 577-84, 2009.
Article in English | MEDLINE | ID: mdl-19296407

ABSTRACT

Earlier studies showed that human lens ALDH1A1 plays a critical role in protection against oxidative stress-induced cytotoxicity in human lens epithelial cells (HLEC), and opacification of rat and mouse lens. The complete coding sequence of ALDH1A1 was cloned from human lens cDNA library by using PCR methods and expressed it in Escherichia coli. The cloned human lens ALDH1A1 cDNA encodes a 501-amino-acid protein (molecular mass = 54.8 kD) that is 100% identical to human liver ALDH1A1 and shares significant identity with the same isozyme from other tissues and species. The purified recombinant human lens ALDH1A1 exhibited optimal catalytic activity at pH 8 and preferred NAD(+) as cofactor and specifically catalyzed the oxidation of toxic lipid aldehydes such as 4-hydroxynonenal (HNE; K(m) = 4.8 microM) and malonaldehyde (K(m) MDA = 3.5 microM). Citral, disulfiram, and cyanamide were found to inhibit human lens ALDH1A1 at IC50 values of 55, 101, and 22610 microM, respectively, whereas diethylstilbestrol (DES) was found to be an activator (EC(50), 1.3 microM). Further, modification of recombinant human lens ALDH1A1 with nitric oxide donors such as S-nitroso-N-acetylpenicillamine (SNAP) and S-nitrosoglutathione (GSNO) significantly inhibited the enzyme activity. It therefore appears that activation of ALDH1A1, which efficiently catalyzes the detoxification of lipid-derived toxic aldehydes, and/or prevention of its oxidative modification may be novel therapeutic interventions against oxidative stress-induced lens pathologies.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Cataract/enzymology , Lens, Crystalline/enzymology , Oxidative Stress , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase 1 Family , Cataract/etiology , Humans , Oxidation-Reduction , Recombinant Proteins/metabolism , Retinal Dehydrogenase
16.
Biochemistry ; 45(40): 12253-64, 2006 Oct 10.
Article in English | MEDLINE | ID: mdl-17014078

ABSTRACT

The Fas (apo/CD95) receptor which belongs to the TNF-alpha family is a transmembrane protein involved in the signaling for apoptosis through the extrinsic pathway. During this study, we have examined a correlation between intracellular levels of 4-HNE and expression of Fas in human lens epithelial (HLE B-3) cells. Our results show that in HLE B-3 cells, Fas is induced by 4-HNE in a concentration- and time-dependent manner, and it is accompanied by the activation of JNK, caspase 3, and the onset of apoptosis. Fas induction and activation of JNK are also observed in various tissues of mGsta4 null mice which have elevated levels of 4-HNE. Conversely, when 4-HNE is depleted in HLE B-3 cells by a transient transfection with hGSTA4, Fas expression is suppressed. However, upon the cessation of hGSTA4 expression in these transiently transfected cells, Fas and 4-HNE return to their basal levels. Fas-deficient transformed HLE B-3 cells stably transfected with hGSTA4 show remarkable resistance to apoptosis. Also, the wild-type HLE B-3 cells in which Fas is partially depleted by siRNA acquire resistance to 4-HNE-induced apoptosis, suggesting an at least partial role of Fas in 4-HNE-induced apoptosis in HLE B-3 cells. We also demonstrate that during 4-HNE-induced apoptosis of HLE B-3 cells, Daxx is induced and it binds to Fas. Together, these results show an important role of 4-HNE in regulation of the expression and functions of Fas.


Subject(s)
Aldehydes/metabolism , Apoptosis/physiology , Signal Transduction/physiology , fas Receptor/genetics , Aldehydes/pharmacology , Animals , Caspase 3 , Caspases/metabolism , Cell Transformation, Viral , Cells, Cultured , Down-Regulation , Gene Expression Regulation/drug effects , Humans , Lens, Crystalline , MAP Kinase Kinase 4/metabolism , Mice , fas Receptor/biosynthesis
17.
Exp Eye Res ; 83(2): 408-16, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16631166

ABSTRACT

Aldehydes are widespread environmental and industrial compounds, which cause cytotoxicity, tissue damage, mutagenicity, and carcinogenicity leading to various disease conditions such as cardiovascular, bronchial, and visual complications. We have shown earlier that aldose reductase (AR) besides reducing glucose to sorbitol, efficiently reduces various toxic lipid-derived aldehydes, generated under oxidative stress, with K(m) in the physiological range. We have identified the role of AR in the prevention of various lipid aldehyde-induced cytotoxic signals leading to apoptosis in human lens epithelial cells (HLEC). HLEC were cultured without or with AR inhibitors followed by addition of various saturated and unsaturated lipid aldehydes with a carbon chain length varying from C3 to C10. The cell viability was assessed by cell counts and MTT assay, and apoptosis was measured by evaluating nucleosomal degradation and caspase-3 activation using specific ELISA kits. Although all the aldehydes caused apoptosis of HLEC, the unsaturated aldehydes were more toxic than saturated aldehydes. Inhibition of AR by sorbinil potentiated while the over-expression of AR prevented the apoptosis induced by various lipid aldehydes. AR over-expression also prevented the lipid aldehyde-induced activation of caspase-3, MAPK, JNK and the expression of Bcl-2 family of proteins in HLEC. The results indicate that the lipid aldehydes generated under oxidative stress are cytotoxic to HLEC leading to apoptosis and that the reduction of lipid aldehydes by AR would prevent it.


Subject(s)
Aldehyde Reductase/metabolism , Aldehydes/toxicity , Environmental Pollutants/toxicity , Lens, Crystalline/enzymology , Aldehyde Reductase/antagonists & inhibitors , Apoptosis/drug effects , Caspase 3 , Caspases/metabolism , Cell Count/methods , Cell Survival/drug effects , Cells, Cultured , Enzyme Activation , Enzyme Inhibitors/metabolism , Epithelial Cells/enzymology , Gene Expression Regulation/genetics , Humans , Imidazolidines/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Oxidative Stress , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Invest Ophthalmol Vis Sci ; 46(1): 259-67, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15623782

ABSTRACT

PURPOSE: 4-Hydroxynonenal (HNE), a metastable lipid peroxidation product, is highly toxic to various cell types if not detoxified. Because of its constant exposure to light, the ocular lens continuously generates reactive oxygen species which, under conditions of oxidative stress, may lead to excessive lipid peroxidation and consequent formation of lipid-derived aldehydes (LDAs) such as HNE. The contribution of various isozymes of aldehyde dehydrogenase (ALDH) to the oxidation of LDAs has never been systematically investigated in the lens. The present study was undertaken to ascertain the role of ALDH1A1 and -3A1 in HNE metabolism and HNE-induced toxicity in cultured human lens epithelial cells (HLECs) and in rat and mouse lenses. METHODS: The metabolism of 3H-HNE was studied in ALDH3A1-knockout mouse lens and in HLECs transfected with ALDH1A1- or -3A1-specific antisense RNA and short interfering (Si)RNA. Appropriate controls were used, including wild-type mouse lens, scrambled oligonucleotides, and a transfection reagent. Transfected HLECs were exposed to oxidative stress (Fenton reaction) or HNE (30 microM) for 3 hours. Toxicity parameters, such as cell viability, apoptosis, and protein-HNE adducts and oxidation of exogenously added 3H-HNE were measured. Rat lenses were transfected with the SiRNA specific to ALDH1A1, and oxidation of 3H-HNE and the susceptibility of the transfected lenses to oxidation-induced opacification were measured. RESULTS: Rat lenses transfected with ALDH1A1-specific SiRNA, or cultured in the presence of the ALDH inhibitor cyanamide/disulfiram and subjected to oxidative stress displayed accelerated loss of transparency and a diminished capacity to oxidize HNE. Similarly, inhibition of ALDH1A1 in HLECs by ALDH1A1-specific antisense RNA or SiRNA was associated with decreased oxidation of 3H-HNE and increased susceptibility of the cells to oxidative damage, including apoptosis. Furthermore, 3H-HNE metabolism and HNE-induced toxicity were not affected in ALDH3A1-specific SiRNA- or antisense RNA-treated rat lenses, HLECs, or ALDH3A1-null mouse lenses. CONCLUSIONS: The results suggest that, under oxidative stress, HNE produced in the lens epithelium can cause toxicity and thus contribute to oxidation-induced cataractogenesis. Furthermore, the studies indicate that ALDH1A1 is a critical isozyme for maintaining clarity in human, rat, and mouse lenses.


Subject(s)
Aldehyde Dehydrogenase/physiology , Cataract/enzymology , Epithelial Cells/enzymology , Lens, Crystalline/enzymology , Oxidative Stress , Aldehyde Dehydrogenase/antagonists & inhibitors , Aldehyde Dehydrogenase/genetics , Aldehydes/metabolism , Aldehydes/toxicity , Animals , Apoptosis/drug effects , Cataract/pathology , Cataract/prevention & control , Cell Survival/drug effects , Cells, Cultured , Chromatography, High Pressure Liquid , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Gas Chromatography-Mass Spectrometry , Humans , Inactivation, Metabolic , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/physiology , Lens, Crystalline/drug effects , Mice , Mice, Knockout , RNA, Antisense/genetics , RNA, Small Interfering/genetics , Rabbits , Rats , Reverse Transcriptase Polymerase Chain Reaction , Transfection
19.
Invest Ophthalmol Vis Sci ; 44(6): 2675-82, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12766072

ABSTRACT

PURPOSE: An earlier study showed that 4-hydroxynonenal (HNE), formed as a result of increased lipid peroxidation in oxidative stress, causes loss of lens transparency. To determine how HNE is detoxified in ocular tissues, its metabolism in cultured human lens epithelial cells (HLECs) as well as rat lens was investigated. METHODS: Rat lens or HLECs were incubated with 30 nmol (5 x 10(5) cpm/ micromol) of HNE in 2 mL Krebs-Hansleit buffer for 1 hour at 37 degrees C. The medium, after ultrafiltration was analyzed by high performance liquid chromatography (HPLC), using a C-18 reversed-phase column. The metabolites were separated by using a gradient consisting of solvent A (0.1% aqueous trifluoroacetic acid) and solvent B (100% acetonitrile) at a flow rate of 1 mL/min. Fractions containing radioactivity were pooled and analyzed using electrospray ionization mass spectroscopy (ESI-MS) or gas chromatography-chemical ionization mass spectroscopy (GC/CI-MS). RESULTS: On HPLC, the incubation media from cultured lens and HLECs separated into three major radioactive peaks. Peak I of the HLECs and lens treated with HNE was identified to be a mixture of glutathione (GS) conjugates of HNE and 1,4-dihydroxy-2-nonene (DHN). The identity of the conjugates was confirmed by ESI-MS. Based on the retention times, peaks II, and III were assigned to 4-hydroxy-2-nonenoic acid (HNA) and unmetabolized HNE, respectively. The identities of HNA and HNE were confirmed by spiking the tissue extracts with synthetic metabolites and finally by GC/CI-MS. Sorbinil, an aldose reductase (AR) inhibitor, attenuated GS-DHN levels and cyanamide, an aldehyde dehydrogenase inhibitor, decreased formation of HNA. CONCLUSIONS: The results show that the major metabolic transformation of HNE in rat lens and HLECs involves conjugation with GS and oxidation to HNA. The GS-HNE conjugate is reduced to GS-DHN by AR. Thus, under normal physiological conditions, the lens has multiple routes to detoxify HNE. However, oxidative stress may overwhelm the metabolic capacity of the lens to detoxify HNE and lead to formation of cataract.


Subject(s)
Aldehydes/metabolism , Epithelial Cells/metabolism , Lens, Crystalline/metabolism , Alkenes/metabolism , Animals , Cells, Cultured , Chromatography, High Pressure Liquid , Gas Chromatography-Mass Spectrometry , Glutathione/metabolism , Humans , Lens, Crystalline/cytology , Lipid Metabolism , Male , Oxidation-Reduction , Oxidative Stress , Rats , Rats, Sprague-Dawley , Spectrometry, Mass, Electrospray Ionization
20.
Invest Ophthalmol Vis Sci ; 43(2): 434-45, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11818388

ABSTRACT

PURPOSE: To investigate the physiological role of two major alpha-class glutathione S-transferases (GSTs), hGSTA1-1 and hGSTA2-2 in protection against oxidative stress and lipid peroxidation (LPO) in human lens epithelial (HLE B-3) cells. METHODS: Total GSTs were purified from HLE B-3 cells by glutathione (GSH)-affinity chromatography and characterized by Western blot analysis, isoelectric focusing, and kinetic studies. The relative contributions of the alpha-class GSTs and the Se-dependent glutathione peroxidase (GPx)-1 in GSH-dependent reduction of phospholipid hydroperoxide (PL-OOH) were quantitated through immunoprecipitation studies using separately the specific polyclonal antibodies against human alpha-class GSTs and GPx-1. HLE B-3 cell membranes were prepared, peroxidized, and used to examine whether hGSTA1-1 and hGSTA2-2 catalyzes the reduction of membrane PL-OOH in situ using the microiodometric and spectrophotometric assays. The protective effects of the alpha-class GSTs against H2O2- and naphthalene-induced LPO and apoptosis were examined by transfecting HLE B-3 cells with cDNAs of hGSTA1 and hGSTA2. RESULTS. HLE B-3 cells expressed only the alpha and pi class GSTs. The Michaelis-Menten constant (k(m)) and turnover number (k(cat)) of purified total GSTs toward phosphatidylcholine hydroperoxide (PC-OOH) were found to be 30 +/- 4 microM and 1.95 +/- 0.26 seconds, respectively. The alpha-class GSTs accounted for approximately 65% of the total GPx activity of HLE B-3 cells toward PC-OOH. Our results demonstrate for the first time that hGSTA1-1 and hGSTA2-2 effectively catalyzed GSH-dependent reduction of membrane PL-OOH in situ in HLE B-3 cells. Transfection with hGSTA1 or hGSTA2 protected these cells from H2O2- and naphthalene-induced LPO and attenuated H2O2- and naphthalene-induced apoptosis through inhibiting caspase 3 activation. CONCLUSIONS: These results demonstrate that the alpha-class GSTs hGSTA1-1 and hGSTA2-2 play a major role as antioxidant enzymes and are the main determinants of the levels of LPO caused by oxidative stress in human lens epithelial cells.


Subject(s)
Apoptosis/drug effects , Epithelial Cells/enzymology , Glutathione Transferase/physiology , Hydrogen Peroxide/toxicity , Isoenzymes/physiology , Lens, Crystalline/enzymology , Lipid Peroxidation/drug effects , Naphthalenes/toxicity , Oxidative Stress , Animals , Blotting, Western , Caspase 3 , Caspases/metabolism , Cells, Cultured , Cytoprotection , Epithelial Cells/drug effects , Epithelial Cells/pathology , Glutathione Peroxidase/metabolism , Glutathione Transferase/isolation & purification , Humans , Isoenzymes/isolation & purification , Lens, Crystalline/drug effects , Lens, Crystalline/pathology , Lipid Peroxides/metabolism , Malondialdehyde/metabolism , Precipitin Tests , Rabbits , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...