Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Radiat Res ; 201(1): 55-70, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38059553

ABSTRACT

Currently, no radioprotectors have been approved to mitigate hematopoietic injury after exposure to ionizing radiation. Acute ionizing radiation results in damage to both hematopoietic and immune system cells. Pre-exposure prophylactic agents are needed for first responders and military personnel. In this study, the ability of gamma-tocotrienol (GT3), a promising radioprotector and antioxidant, to ameliorate partial-body radiation-induced damage to the hematopoietic compartment was evaluated in a nonhuman primate (NHP) model. A total of 15 rhesus NHPs were divided into two groups, and were administered either GT3 or vehicle 24 h prior to 4 or 5.8 Gy partial-body irradiation (PBI), with 5% bone marrow (BM) sparing. Each group consisted of four NHPs, apart from the vehicle-treated group exposed to 5.8 Gy, which had only three NHPs. BM samples were collected 8 days prior to irradiation in addition to 2, 7, 14, and 30 days postirradiation. To assess the clonogenic ability of hematopoietic stem and progenitor cells (HSPCs), colony forming unit (CFU) assays were performed, and lymphoid cells were immunophenotyped using flow cytometry. As a result of GT3 treatment, an increase in HSPC function was evident by an increased recovery of CFU-granulocyte macrophages (CFU-GM). Additionally, GT3 treatment was shown to increase the percentage of CD34+ cells, including T and NK-cell subsets. Our data further affirm GT3's role in hematopoietic recovery and suggest the need for its further development as a prophylactic radiation medical countermeasure.


Subject(s)
Chromans , Radiation-Protective Agents , Animals , Macaca mulatta , Radiation-Protective Agents/pharmacology , Vitamin E/pharmacology , Bone Marrow/radiation effects
2.
Int J Mol Sci ; 23(24)2022 Dec 18.
Article in English | MEDLINE | ID: mdl-36555814

ABSTRACT

Radiation exposure causes acute damage to hematopoietic and immune cells. To date, there are no radioprotectors available to mitigate hematopoietic injury after radiation exposure. Gamma-tocotrienol (GT3) has demonstrated promising radioprotective efficacy in the mouse and nonhuman primate (NHP) models. We determined GT3-mediated hematopoietic recovery in total-body irradiated (TBI) NHPs. Sixteen rhesus macaques divided into two groups received either vehicle or GT3, 24 h prior to TBI. Four animals in each treatment group were exposed to either 4 or 5.8 Gy TBI. Flow cytometry was used to immunophenotype the bone marrow (BM) lymphoid cell populations, while clonogenic ability of hematopoietic stem cells (HSCs) was assessed by colony forming unit (CFU) assays on day 8 prior to irradiation and days 2, 7, 14, and 30 post-irradiation. Both radiation doses showed significant changes in the frequencies of B and T-cell subsets, including the self-renewable capacity of HSCs. Importantly, GT3 accelerated the recovery in CD34+ cells, increased HSC function as shown by improved recovery of CFU-granulocyte macrophages (CFU-GM) and burst-forming units erythroid (B-FUE), and aided the recovery of circulating neutrophils and platelets. These data elucidate the role of GT3 in hematopoietic recovery, which should be explored as a potential medical countermeasure to mitigate radiation-induced injury to the hematopoietic system.


Subject(s)
Hematopoietic Stem Cells , Vitamin E , Mice , Animals , Macaca mulatta , Vitamin E/pharmacology , Chromans/pharmacology , Whole-Body Irradiation
3.
Antioxidants (Basel) ; 11(10)2022 Sep 25.
Article in English | MEDLINE | ID: mdl-36290618

ABSTRACT

Exposure to high doses of radiation, accidental or therapeutic, often results in gastrointestinal (GI) injury. To date, there are no therapies available to mitigate GI injury after radiation exposure. Gamma-tocotrienol (GT3) is a promising radioprotector under investigation in nonhuman primates (NHP). We have shown that GT3 has radioprotective function in intestinal epithelial and crypt cells in NHPs exposed to 12 Gy total-body irradiation (TBI). Here, we determined GT3 potential in accelerating the GI recovery in partial-body irradiated (PBI) NHPs using X-rays, sparing 5% bone marrow. Sixteen rhesus macaques were treated with either vehicle or GT3 24 h prior to 12 Gy PBI. Structural injuries and crypt survival were examined in proximal jejunum on days 4 and 7. Plasma citrulline was assessed using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Crypt cell proliferation and apoptotic cell death were evaluated using Ki-67 and TUNEL staining. PBI significantly decreased mucosal surface area and reduced villous height. Interestingly, GT3 increased crypt survival and enhanced stem cell proliferation at day 4; however, the effects seemed to be minimized by day 7. GT3 did not ameliorate a radiation-induced decrease in citrulline levels. These data suggest that X-rays induce severe intestinal injury post-PBI and that GT3 has minimal radioprotective effect in this novel model.

4.
J Clin Invest ; 132(18)2022 09 15.
Article in English | MEDLINE | ID: mdl-35881476

ABSTRACT

Osteolytic bone disease is a hallmark of multiple myeloma (MM). A significant fraction (~20%) of MM patients do not develop osteolytic lesions (OLs). The molecular basis for the absence of bone disease in MM is not understood. We combined PET-CT and gene expression profiling (GEP) of purified BM CD138+ MM cells from 512 newly diagnosed MM patients to reveal that elevated expression of cystatin M/E (CST6) was significantly associated with the absence of OL in MM. An enzyme-linked immunosorbent assay revealed a strong correlation between CST6 levels in BM serum/plasma and CST6 mRNA expression. Both recombinant CST6 protein and BM serum from patients with high CST6 significantly inhibited the activity of the osteoclast-specific protease cathepsin K and blocked osteoclast differentiation and function. Recombinant CST6 inhibited bone destruction in ex vivo and in vivo myeloma models. Single-cell RNA-Seq showed that CST6 attenuates polarization of monocytes to osteoclast precursors. Furthermore, CST6 protein blocks osteoclast differentiation by suppressing cathepsin-mediated cleavage of NF-κB/p100 and TRAF3 following RANKL stimulation. Secretion by MM cells of CST6, an inhibitor of osteoclast differentiation and function, suppresses osteolytic bone disease in MM and probably other diseases associated with osteoclast-mediated bone loss.


Subject(s)
Bone Resorption , Multiple Myeloma , Osteolysis , Bone Resorption/genetics , Bone Resorption/metabolism , Cell Differentiation/physiology , Cystatin M/metabolism , Humans , Multiple Myeloma/complications , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Osteoclasts/metabolism , Osteolysis/genetics , Osteolysis/metabolism , Positron Emission Tomography Computed Tomography , RANK Ligand/genetics , RANK Ligand/metabolism , TNF Receptor-Associated Factor 3/metabolism
5.
Int J Mol Sci ; 23(9)2022 Apr 22.
Article in English | MEDLINE | ID: mdl-35563033

ABSTRACT

The gastrointestinal (GI) system is highly susceptible to irradiation. Currently, there is no Food and Drug Administration (FDA)-approved medical countermeasures for GI radiation injury. The vitamin E analog gamma-tocotrienol (GT3) is a promising radioprotector in mice and nonhuman primates (NHP). We evaluated GT3-mediated GI recovery in total-body irradiated (TBI) NHPs. Sixteen rhesus macaques were divided into two groups; eight received vehicle and eight GT3 24 h prior to 12 Gy TBI. Proximal jejunum was assessed for structural injuries and crypt survival on day 4 and 7. Apoptotic cell death and crypt cell proliferation were assessed with TUNEL and Ki-67 immunostaining. Irradiation induced significant shortening of the villi and reduced mucosal surface area. GT3 induced an increase in crypt depth at day 7, suggesting that more stem cells survived and proliferated after irradiation. GT3 did not influence crypt survival after irradiation. GT3 treatment caused a significant decline in TUNEL-positive cells at both day 4 (p < 0.03) and 7 (p < 0.0003). Importantly, GT3 induced a significant increase in Ki-67-positive cells at day 7 (p < 0.05). These data suggest that GT3 has radioprotective function in intestinal epithelial and crypt cells. GT3 should be further explored as a prophylactic medical countermeasure for radiation-induced GI injury.


Subject(s)
Acute Radiation Syndrome , Chromans , Radiation-Protective Agents , Vitamin E , Acute Radiation Syndrome/drug therapy , Acute Radiation Syndrome/prevention & control , Animals , Chromans/therapeutic use , Disease Models, Animal , Intestines/pathology , Intestines/radiation effects , Ki-67 Antigen , Macaca mulatta , Radiation-Protective Agents/pharmacology , Radiation-Protective Agents/therapeutic use , Vitamin E/analogs & derivatives , Vitamin E/therapeutic use
6.
PLoS One ; 8(1): e54610, 2013.
Article in English | MEDLINE | ID: mdl-23372742

ABSTRACT

Multicentric Castleman Disease is largely driven by increased signaling in the pathway for the plasma cell growth factor interleukin-6. We hypothesized that interleukin-6/interleukin-6 receptor/gp130 polymorphisms contribute to increased interleukin-6 and/or other components of the interleukin-6 signaling pathway in HIV-negative Castleman Disease patients. The study group was composed of 58 patients and 50 healthy donors of a similar racial/ethnic profile. Of seven polymorphisms chosen for analysis, we observed an increased frequency between patients and controls of the minor allele of interleukin-6 receptor polymorphism rs4537545, which is in linkage disequilibrium with interleukin-6 receptor polymorphism rs2228145. Further, individuals possessing at least one copy of the minor allele of either polymorphism expressed higher levels of soluble interleukin-6 receptor. These elevated interleukin-6 receptor levels may contribute to increased interleukin-6 activity through the trans-signaling pathway. These data suggest that interleukin-6 receptor polymorphism may be a contributing factor in Castleman Disease, and further research is warranted.


Subject(s)
Castleman Disease/genetics , Castleman Disease/metabolism , Polymorphism, Single Nucleotide , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism , Alleles , Case-Control Studies , Female , Gene Frequency , Genotype , Humans , Interleukin-6/blood , Interleukin-6/genetics , Male
7.
Haematologica ; 97(9): 1348-56, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22419581

ABSTRACT

BACKGROUND: Patients with gene expression profiling-defined high-risk myeloma in relapse have poor outcomes with current therapies. We tested whether natural killer cells expanded by co-culture with K562 cells transfected with 41BBL and membrane-bound interleukin-15 could kill myeloma cells with a high-risk gene expression profile in vitro and in a unique model which recapitulates human myeloma. DESIGN AND METHODS: OPM2 and high-risk primary myeloma tumors were grown in human fetal bone implanted into non-obese diabetic severe combined immunodeficiency mice with a deficient interleukin-2 receptor gamma chain. These mice are devoid of endogenous natural killer and T-cell activity and were used to determine whether adoptively transferred expanded natural killer cells could inhibit myeloma growth and myeloma-associated bone destruction. RESULTS: Natural killer cells from healthy donors and myeloma patients expanded a median of 804- and 351-fold, respectively, without significant T-cell expansion. Expanded natural killer cells killed both allogeneic and autologous primary myeloma cells avidly via a perforin-mediated mechanism in which the activating receptor NKG2D, natural cytotoxicity receptors, and DNAX-accessory molecule-1 played a central role. Adoptive transfer of expanded natural killer cells inhibited the growth of established OPM2 and high-risk primary myeloma tumors grown in the murine model. The transferred, expanded natural killer cells proliferated in vivo in an interleukin-2 dose-dependent fashion, persisted up to 4 weeks, were readily detectable in the human bone, inhibited myeloma growth and protected bone from myeloma-induced osteolysis. CONCLUSIONS: These studies provide the rationale for testing expanded natural killer cells in humans.


Subject(s)
Cytotoxicity, Immunologic/immunology , Immunotherapy, Adoptive , Killer Cells, Natural/immunology , Multiple Myeloma/therapy , T-Lymphocytes/immunology , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Coculture Techniques , Flow Cytometry , Humans , Immunoenzyme Techniques , Interleukin Receptor Common gamma Subunit/genetics , Interleukin-2/metabolism , Killer Cells, Natural/metabolism , Lymphocyte Activation , Mice , Mice, Inbred NOD , Mice, SCID , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Osteolysis , T-Lymphocytes/metabolism , Tumor Cells, Cultured
8.
Neotrop. ichthyol ; 8(4): 877-884, 2010. ilus, tab
Article in English | LILACS | ID: lil-571579

ABSTRACT

The structural organization and histo-cytochemical features of dorsal skin of Ancistrus dolichopterus (acari bodo) are the main focus of this work. The epidermis, dermis and subcutis are the principal layers of the skin. The epidermis mainly consists of epithelial and mucous cells. Interspersed between them are lymphocytes, pigment cells, eosinophilic granular cells (EGC), and the taste buds as sensory structures. The high number of EGCs is implicated in general and specific immunological defense from pathogenic bacteria and multicellular parasites. The epithelial cells and mucous cells contain glycoproteins with oxidizable vicinal diols, carboxyl groups and O-sulphate esters and their high secretory activity is correlated with the bottom dwelling habit of this species. A thick stratum laxum contains overlapping osteoderms bearing denticles, and the stratum compactum make the integument thicker to help the fish in negative buoyancy for maneuvering near the bottom and protection. The entire body surface is covered by conical, backwardly directed denticles. These are composed of a dentine cone, surrounding a pulp cavity with the top covered by mineralized cap, and are the true homologues of teeth. These structures provide effective protection from abrasion and enemies. These structural peculiarities and histochemical features indicate additional physiological role of the skin of A. dolichopterus.


A organização estrutural e aspectos histo-citiquímicos da pele dorsal de Ancistrus dolichopterus (acari-bodó) são os principais alvos do presente estudo. A epiderme, a derme e a hipoderme são as principais camadas da pele. A epiderme consiste principalmente de células epiteliais e mucosas. Intercalados entre elas estão os linfócitos, as células pigmentares, as células granulares eosinofílicas (CGE), e as papilas gustativas como estruturas sensoriais. Um grande número de CGEs está relacionado em geral com a defesa imunológica específica de bactérias patogênicas e parasitas multicelulares. As células epiteliais e as células mucosas contem glicoproteínas com grupos diol oxidáveis, grupos carboxilas e ésteres O-sulfatados sendo que sua alta atividade secretória está correlacionada com o comportamento bentônico, de fundo, dessa espécie. Um espesso stratum laxum contém osteodermos sobrepostos, parecidos com dentículos, e o stratum compactum que torna o tegumento mais espesso, contribuindo com a flutuação negativa necessária ao movimento perto do fundo e proteção. Toda a superfície do corpo é coberta por dentículos cônicos retrodirecionados. Esses dentículos são compostos por um cone de dentina, envolvendo uma cavidade pulpar e com o ápice coberto por uma capa mineralizada, verdadeiros homólogos dos dentes. Essas estruturas oferecem efetiva proteção contra abrasão e oponentes. Essas peculiaridades estruturais e aspectos histoquímicos sugerem a existência de uma função fisiológica adicional para a pele de A. dolichopterus.


Subject(s)
Animals , Dental Pulp Calcification/veterinary , Catfishes/anatomy & histology
9.
Mol Cancer Ther ; 8(9): 2616-24, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19723891

ABSTRACT

Monoclonal antibody (mAb) therapy for multiple myeloma, a malignancy of plasma cells, has not been clinically efficacious in part due to a lack of appropriate targets. We recently reported that the cell surface glycoprotein CS1 (CD2 subset 1, CRACC, SLAMF7, CD319) was highly and universally expressed on myeloma cells while having restricted expression in normal tissues. Elotuzumab (formerly known as HuLuc63), a humanized mAb targeting CS1, is currently in a phase I clinical trial in relapsed/refractory myeloma. In this report we investigated whether the activity of elotuzumab could be enhanced by bortezomib, a reversible proteasome inhibitor with significant activity in myeloma. We first showed that elotuzumab could induce patient-derived myeloma cell killing within the bone marrow microenvironment using a SCID-hu mouse model. We next showed that CS1 gene and cell surface protein expression persisted on myeloma patient-derived plasma cells collected after bortezomib administration. In vitro bortezomib pretreatment of myeloma targets significantly enhanced elotuzumab-mediated antibody-dependent cell-mediated cytotoxicity, both for OPM2 myeloma cells using natural killer or peripheral blood mononuclear cells from healthy donors and for primary myeloma cells using autologous natural killer effector cells. In an OPM2 myeloma xenograft model, elotuzumab in combination with bortezomib exhibited significantly enhanced in vivo antitumor activity. These findings provide the rationale for a clinical trial combining elotuzumab and bortezomib, which will test the hypothesis that combining both drugs would result in enhanced immune lysis of myeloma by elotuzumab and direct targeting of myeloma by bortezomib.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Boronic Acids/therapeutic use , Membrane Glycoproteins/immunology , Multiple Myeloma/drug therapy , Pyrazines/therapeutic use , Animals , Antibodies, Monoclonal, Humanized , Antibody-Dependent Cell Cytotoxicity , Apoptosis/drug effects , Bortezomib , Case-Control Studies , Female , Flow Cytometry , Gene Expression Profiling , Humans , Immunohistochemistry , Membrane Glycoproteins/genetics , Mice , Mice, SCID , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Transplantation, Heterologous
10.
Br J Haematol ; 143(5): 641-53, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18950462

ABSTRACT

Killer immunoglobulin-like receptor (KIR)-ligand mismatched natural killer (NK) cells play a key role in achieving durable remission after haplo-identical transplantation for acute myeloid leukaemia. We investigated the feasibility of transfusing haplo-identical, T-cell depleted, KIR-ligand mismatched NK cells, after conditioning therapy with melphalan and fludarabine, to patients with advanced multiple myeloma (MM) followed by delayed rescue with autologous stem cells. No graft-versus-host disease or failure of autologous stem cells to engraft was observed. There was significant variation in the number of allo-reactive NK cells transfused. However, all NK products containing allo-reactive NK cells killed the NK cell target K562, the MM cell line U266, and recipient MM cells when available. Post NK cell infusion there was a rise in endogenous interleukin-15 accompanied by increasing donor chimaerism. Donor chimaerism was eventually lost, which correlated with the emergence of potent host anti-donor responses indicating that the immunosuppressive properties of the conditioning regimen require further optimization. Further, blocking of inhibitory KIR-ligands with anti-human leucocyte antigen antibody substantially enhanced killing of MM cells thus highlighting the potential for modulating NK/MM cell interaction. Encouragingly, 50% of patients achieved (near) complete remission. These data set the stage for future studies of KIR-ligand mismatched NK cell therapy in the autologous setting.


Subject(s)
Hematopoietic Stem Cell Transplantation , Immunotherapy, Adoptive/methods , Killer Cells, Natural/transplantation , Multiple Myeloma/therapy , Receptors, KIR/immunology , Adult , Aged , Cell Line , Cytotoxicity Tests, Immunologic , Female , Haplotypes , Humans , Killer Cells, Natural/immunology , Ligands , Male , Middle Aged , Multiple Myeloma/immunology , Recurrence , Transplantation, Autologous , Treatment Outcome
11.
Alcohol ; 42(1): 29-36, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18249267

ABSTRACT

Oxidative stress is a candidate mechanism for ethanol neuropathology in fetal alcohol spectrum disorders. Oxidative stress often involves production of reactive oxygen species (ROS), deterioration of the mitochondrial membrane potential (MMP), and cell death. Previous studies have produced conflicting results regarding the role of oxidative stress and the benefit of antioxidants in ethanol neuropathology in the developing brain. This study investigated the hypothesis that ethanol neurotoxicity involves production of ROS with negative downstream consequences for MMP and neuron survival. This was modeled in neonatal rats at postnatal day 4 (P4) and P14. It is well established that granule neurons in the rat cerebellar cortex are more vulnerable to ethanol neurotoxicity on P4 than at later ages. Thus, it was hypothesized that ethanol produces more oxidative stress and its negative consequences on P4 than on P14. A novel experimental approach was used in which ethanol was administered to animals in vivo (gavage 6g/kg), granule neurons were isolated 2-24h post-treatment, and ROS production and relative MMP were immediately assessed in the viable cells. Cells were also placed in culture and survival was measured 24h later. The results revealed that ethanol did not induce granule cells to produce ROS, cause deterioration of neuronal MMP, or cause neuron death when compared to vehicle controls. Further, granule neurons from neither P4 nor P14 animals mounted an oxidative response to ethanol. These findings do not support the hypothesis that oxidative stress is obligate to granule neuron death after ethanol exposure in the neonatal rat brain. Other investigators have reached a similar conclusion using either brain homogenates or cell cultures. In this context, it is likely that oxidative stress is not the sole and perhaps not the principal mechanism of ethanol neurotoxicity for cerebellar granule neurons during this stage of brain development.


Subject(s)
Cerebellum/drug effects , Ethanol/toxicity , Oxidative Stress , Animals , Animals, Newborn , Biomarkers , Matrix Metalloproteinases/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
12.
Blood ; 111(3): 1309-17, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-17947507

ABSTRACT

Human leukocyte antigen class I molecules expressed by tumor cells play a central role in the regulation of natural killer (NK) cell-mediated immune responses. The proteasome inhibitor bortezomib has demonstrated significant activity in multiple myeloma (MM). We hypothesized that treatment of MM with bortezomib results in the reduction of cell-surface expression of class I and thereby sensitizes MM to NK cell-mediated lysis. Here we report that bortezomib down-regulates class I in a time- and dose-dependent fashion on all MM cell lines and patient MM cells tested. Downregulation of class I can also be induced in vivo after a single dose of 1.0 mg/m(2) bortezomib. Bortezomib significantly enhances the sensitivity of patient myeloma to allogeneic and autologous NK cell-mediated lysis. Further, the level of decrease in class I expression correlates with increased susceptibility to lysis by NK cells. Clinically relevant bortezomib concentrations do not affect NK-cell function. Our findings have clear therapeutic implications for MM and other NK cell-sensitive malignancies in the context of both allogeneic and autologous adoptively transferred NK cells.


Subject(s)
Boronic Acids/pharmacology , Cell Membrane/drug effects , Down-Regulation/drug effects , Histocompatibility Antigens Class I/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Multiple Myeloma/immunology , Pyrazines/pharmacology , Bortezomib , Cell Membrane/immunology , Cell Survival/drug effects , Humans , Multiple Myeloma/pathology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Receptors, KIR/classification , Receptors, KIR/immunology , Sensitivity and Specificity , Tumor Cells, Cultured
13.
J Neuroimmunol ; 171(1-2): 17-28, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16225932

ABSTRACT

Methylmercury (MeHg) causes severe neurological disorders in the central nervous system. This study focused on the effects of MeHg on microglia, macrophage-like cells that reside in the CNS important in neuro-immune interactions. The murine N9 microglial cell line was used in this set of study. MeHg caused reactive oxygen species generation, mitochondrial depolarization and aconitase inactivation, all of which were signs of cellular oxidative stress. MeHg greatly increased microglial IL-6 secretion despite the fact that it severely inhibited protein synthesis. The concentration that caused 50% cell death in 24 h was approximately 9 microM. Pretreatment of microglia with the prostaglandin derivative, 15-deoxy-delta 12, 14-Prostaglandin J2 attenuated MeHg induced cell death. The saving effect did not appear to be mediated through activation of peroxisome proliferator activated receptors (PPAR) since other agonists of these receptors did not prevent MeHg induced microglial death.


Subject(s)
Immunologic Factors/pharmacology , Methylmercury Compounds/toxicity , Microglia/drug effects , Oxidative Stress/drug effects , Prostaglandin D2/analogs & derivatives , Aconitate Hydratase/metabolism , Analysis of Variance , Animals , Cell Line , Cell Survival/drug effects , Cytotoxicity Tests, Immunologic/methods , Dose-Response Relationship, Drug , Drug Interactions , Hydrogen Peroxide/pharmacology , Interleukin-6/metabolism , Membrane Potentials/drug effects , Mice , Mitochondria/drug effects , Models, Biological , Oxidative Stress/physiology , Prostaglandin D2/pharmacology , Reactive Oxygen Species/metabolism , Time Factors
14.
BMC Pharmacol ; 4: 6, 2004 May 18.
Article in English | MEDLINE | ID: mdl-15149553

ABSTRACT

BACKGROUND: With the use of cultured human retinal pigment epithelial cells, we have previously described a number of cellular responses to oxidative stress caused by H2O2. We also demonstrated that the cytotoxicity caused by H2O2 could be prevented by the prostaglandin derivative, 15-deoxy-delta 12, 14-Prostaglandin J2 (15d-PGJ2). RESULTS: Further characterization of the experimental system indicated that the half-life of H2O2 in cultures was ~1 hour. At a fixed H2O2 concentration, the cytotoxicity was dependent on the volume of H2O2 solution used in the culture, such that higher volume caused more cytotoxicity. Most cells were committed to die if the culture was treated for 2 hours with a cytotoxic concentration of H2O2. The prostaglandin derivative, 15d-PGJ2, could prevent oxidative damage caused by t-butyl hydroperoxide, in addition to H2O2. Further studies indicated that both H2O2 and tBH caused an increase in reactive oxygen species and depolarization of mitochondrial membrane potential. Pretreatment of cells with 1 microM 15d-PGJ2 led to a modest decrease in reactive oxygen species generation, and a significant restoration of mitochondrial membrane potential. CONCLUSION: This agent may be used in the future as a pharmacological tool for preventing cellular damage caused by oxidative stress.


Subject(s)
Mitochondria/drug effects , Oxidative Stress/drug effects , Pigment Epithelium of Eye/drug effects , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/pharmacology , Reactive Oxygen Species/metabolism , Cells, Cultured , Humans , Hydrogen Peroxide/metabolism , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mitochondria/physiology , Oxidative Stress/physiology , Pigment Epithelium of Eye/physiology
15.
BMC Ophthalmol ; 3: 5, 2003 Mar 21.
Article in English | MEDLINE | ID: mdl-12659653

ABSTRACT

BACKGROUND: The retina, which is exposed to both sunlight and very high levels of oxygen, is exceptionally rich in polyunsaturated fatty acids, which makes it a favorable environment for the generation of reactive oxygen species. The cytotoxic effects of hydrogen peroxide (H2O2) induced oxidative stress on retinal pigment epithelium were characterized in this study. METHODS: The MTT cell viability assay, Texas-Red phalloidin staining, immunohistochemistry and Western blot analysis were used to assess the effects of oxidative stress on primary human retinal pigment epithelial cell cultures and the ARPE-19 cell line. RESULTS: The treatment of retinal pigment epithelial cells with H2O2 caused a dose-dependent decrease of cellular viability, which was preceded by a significant cytoskeletal rearrangement, activation of the Extracellular signal-Regulated Kinase, lipid peroxidation and nuclear condensation. This cell death was prevented partially by the prostaglandin derivative, 15d-PGJ2 and by the protein kinase inhibitor, AG126. CONCLUSION: 15d-PGJ2 and AG126 may be useful pharmacological tools in the future capable of preventing oxidative stress induced RPE cell death in human ocular diseases.


Subject(s)
Enzyme Inhibitors/pharmacology , Immunologic Factors/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Oxidative Stress , Pigment Epithelium of Eye/drug effects , Prostaglandin D2/pharmacology , Tyrphostins/pharmacology , Actins/metabolism , Aldehydes/metabolism , Blotting, Western , Cell Death , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Fluorescent Antibody Technique, Indirect , Humans , Hydrogen Peroxide/toxicity , Immunoenzyme Techniques , Lipid Peroxidation/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Phosphorylation , Pigment Epithelium of Eye/metabolism , Pigment Epithelium of Eye/pathology , Prostaglandin D2/analogs & derivatives , Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/agonists , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...