Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Nutr ; 150(5): 1100-1108, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32133527

ABSTRACT

BACKGROUND: Radiotherapy inadvertently affects gastrointestinal (GI) epithelial cells, causing intestinal barrier disruption and increased permeability. OBJECTIVE: We examined the effect of amino acid-based oral rehydration solution (AA-ORS) on radiation-induced changes of intestinal barrier function and epithelial tight junctions (TJs) in a randomized experimental study using a total-body irradiation (TBI) mouse model. METHODS: Eight-week-old male Swiss mice received a single-dose TBI (0, 1, 3, or 5 Gy), and subsequent gastric gavage with AA-ORS (threonine, valine, serine, tyrosine, and aspartic acid) or saline for 2 or 6 d. Intestinal barrier function of mouse ileum was characterized by electrophysiological analysis of conductance, anion selectivity, and paracellular permeability [fluorescein isothiocyanate (FITC)-dextran]. Ultrastructural changes of TJs were evaluated by transmission electron microscopy. Membrane protein and mRNA expression of claudin-1, -2, -3, -5, and -7, occludin, and E-cadherin were analyzed with western blot, qPCR, and immunohistochemistry. Nonparametric tests were used to compare treatment-dose differences for each time point. RESULTS: Saline-treated mice had a higher conductance at doses as low as 3 Gy, and as early as 2 d post-TBI compared with 0 Gy (P < 0.001). Paracellular permeability and dilution potential were increased 6 d after 5 Gy TBI (P < 0.001). Conductance decreased with AA-ORS after 2 d in 3-Gy and 5-Gy mice (P < 0.05 and P < 0.001), and on day 6 after 5 Gy TBI (P < 0.001). Anion selectivity and FITC permeability decreased from 0.73 ± 0.02 to 0.61 ± 0.03 pCl/pNa (P < 0.01) and from 2.7 ± 0.1 × 105 to 2.1 ± 0.1 × 105 RFU (P < 0.001) in 5-Gy mice treated with AA-ORS for 6 d compared with saline. Irradiation-induced ultrastructural changes of TJs characterized by decreased electron density and gap formation improved with AA-ORS. Reduced claudin-1, -3, and -7 membrane expression after TBI recovered with AA-ORS within 6 d, whereas claudin-2 decreased indicating restitution of TJ proteins. CONCLUSIONS: Radiation-induced functional and structural disruption of the intestinal barrier in mice is reversed by AA-ORS rendering AA-ORS a potential treatment option in prospective clinical trials in patients with gastrointestinal barrier dysfunction.


Subject(s)
Amino Acids/administration & dosage , Intestines/radiation effects , Rehydration Solutions/chemistry , Rehydration Solutions/pharmacology , Tight Junctions/radiation effects , Animals , Fluid Therapy , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Male , Mice , Permeability , RNA, Messenger , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism
2.
Mol Ther ; 27(3): 493-506, 2019 03 06.
Article in English | MEDLINE | ID: mdl-30713087

ABSTRACT

Nanoparticles (NPs) have been utilized to deliver drugs to the intestinal epithelium in vivo. Moreover, NPs derived from edible plants are less toxic than synthetic NPs. Here, we utilized ginger NP-derived lipid vectors (GDLVs) in a proof-of-concept investigation to test the hypothesis that inhibiting expression of divalent metal-ion transporter 1 (Dmt1) would attenuate iron loading in a mouse model of hereditary hemochromatosis (HH). Initial experiments using duodenal epithelial organ cultures from intestine-specific Dmt1 knockout (KO) (Dmt1int/int) mice in the Ussing chamber established that Dmt1 is the only active iron importer during iron-deficiency anemia. Further, when Dmt1int/int mice were crossed with mice lacking the iron-regulatory hormone, hepcidin (Hepc-/-), iron loading was abolished. Hence, intestinal Dmt1 is required for the excessive iron absorption that typifies HH. Additional experiments established a protocol to produce GDLVs carrying functional Dmt1 small interfering RNAs (siRNAs) and to target these gene delivery vehicles to the duodenal epithelium in vivo (by incorporating folic acid [FA]). When FA-GDLVs carrying Dmt1 siRNA were administered to weanling Hepc-/- mice for 16 days, intestinal Dmt1 mRNA expression was attenuated and tissue iron accumulation was blunted. Oral delivery of functional siRNAs by FA-GDLVs is a suitable therapeutic approach to mitigate iron loading in murine HH.


Subject(s)
Hemochromatosis/metabolism , Hepcidins/metabolism , Nanoparticles/chemistry , Transcription Factors/metabolism , Zingiber officinale , Animals , Female , HEK293 Cells , Hemochromatosis/genetics , Hepcidins/genetics , Humans , Iron/metabolism , Iron, Dietary , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction , Transcription Factors/genetics
3.
Int J Radiat Biol ; 93(12): 1334-1342, 2017 12.
Article in English | MEDLINE | ID: mdl-29063809

ABSTRACT

PURPOSE: To investigate the late effects of thoracic region irradiation (TRI) on mouse body weight. MATERIALS AND METHODS: Female C57BL/6 mice were divided into nonirradiated, 5 Gy total body irradiation, 9 Gy sub-total body irradiation, and 12.5 Gy thoracic region irradiation (TRI) groups. Changes in mouse weight were monitored every other week at similar time points for 12 months. The anatomical characteristics of abdominal visceral fat distribution were recorded, and mitochondrial DNA copy number in the hearts and livers and lipid metabolic signaling in the liver were analyzed. Data were analyzed by one-way analysis of variance and a student's t-test. RESULTS: TRI led to a significant increase (p < .001) in body weight that was dependent on time and individuals [42.1% of mice were overweight (50% increase in body weight) 4 months post-TRI and 100% of mice were overweight at 10 months post-TRI]. Gross anatomical features of abdominal visceral fat distribution and storage in radiation-induced overweight/severely overweight mice were similar to those of high fat diet-induced overweight/severely overweight mice. The mitochondrial genome of heart and liver tissues from overweight/severely overweight mice had significantly (p < .05) decreased functional mitochondrial DNA copy number (ratios decreased from 1 to 0.71 or 0.49, respectively) and significantly (p < .05) increased mitochondrial DNA mutations (ratios increased from 1 to 3.21 or 1.83, respectively). CPT1 and IRS2 lipid metabolic signaling was significantly (p < .05-.01) decreased for both mRNA (fold decrease from 1 to 0.60 or 0.55, respectively) and protein (fold decrease from 1 to 0.62 or 0.19, respectively). CONCLUSIONS: TRI can cause mice to gain weight. These findings indicate that TRI can result in lipid metabolic abnormalities and provide a model to study the factors that result in these abnormalities.


Subject(s)
Gamma Rays/adverse effects , Obesity/etiology , Thorax/radiation effects , Animals , Body Weight/radiation effects , Disease Progression , Female , Genome, Mitochondrial/radiation effects , Lipid Metabolism/radiation effects , Mice , Mice, Inbred C57BL , Obesity/genetics , Obesity/metabolism , Obesity/physiopathology
4.
Adv Exp Med Biol ; 977: 59-65, 2017.
Article in English | MEDLINE | ID: mdl-28685428

ABSTRACT

Nausea and diarrhea are common yet inconsistent side effects of abdominal and pelvic irradiation. Their frequency, chronicity, and severity vary greatly, and the reasons for inter-subject variability are unknown. We studied the potential for radiation-induced changes in amino acid absorption and mucosal barrier function to lead to gastrointestinal toxicity. We found profound and prolonged changes in the absorption and secretion of several electrolytes and nutrients, caused by changes in transporter function, after radiation doses as low as 1 to 3 Gy. After identifying absorbed and non-absorbed amino acids, we demonstrated the role of a beneficial amino acid drink to alleviate radiation-related gastrointestinal symptoms in a mouse model.


Subject(s)
Amino Acids/administration & dosage , Fluid Therapy/methods , Nausea/therapy , Pica/therapy , Radiation Injuries/therapy , Rehydration Solutions/therapeutic use , Amino Acids/pharmacokinetics , Animals , Disease Models, Animal , Electrolytes/pharmacokinetics , Gastrointestinal Absorption , Gastrointestinal Diseases/etiology , Gastrointestinal Diseases/therapy , Glucose/pharmacokinetics , Male , Mice , Nausea/etiology , Pica/pathology , Radiation Injuries/complications , Rehydration Solutions/chemistry
5.
Pflugers Arch ; 469(9): 1093-1105, 2017 09.
Article in English | MEDLINE | ID: mdl-28488023

ABSTRACT

Rotavirus causes severe diarrhea in small children and is typically treated using glucose-containing oral rehydration solutions; however, glucose may have a detrimental impact on these patients, because it increases chloride secretion and presumably water loss. The rotavirus enterotoxin nonstructural protein 4 (NSP4) directly inhibits glucose-mediated sodium absorption. We examined the effects of NSP4 and glucose on sodium and chloride transport in mouse small intestines and Caco-2 cells. Mouse small intestines and Caco-2 cells were incubated with NSP4114-135 in the presence/absence of glucose. Absorption and secretion of sodium and chloride, fluid movement, peak amplitude of intracellular calcium fluorescence, and expression of Ano1 and sodium-glucose cotransporter 1 were assessed. NHE3 activity increased, and chloride secretory activity decreased with age. Net chloride secretion increased, and net sodium absorption decreased in the intestines of 3-week-old mice compared to 8-week-old mice with NSP4. Glucose increased NSP4-stimulated chloride secretion. Glucose increased NSP4-stimulated increase in short-circuit current measurements (I sc) and net chloride secretion. Ano1 cells with siRNA knockdown showed a significant difference in I sc in the presence of NSP4 and glucose without a significant difference in peak calcium fluorescence intracellular when compared to non-silencing (N.S.) cells. The failure of glucose to stimulate significant sodium absorption was likely due to the inhibition of sodium-hydrogen exchange and sodium-glucose cotransport by NSP4. Since glucose enhances intestinal chloride secretion and fails to increase sodium absorption in the presence of NSP4, glucose-based oral rehydration solutions may not be ideal for the management of rotaviral diarrhea.


Subject(s)
Enterotoxins/pharmacology , Glucose/metabolism , Intestinal Mucosa/metabolism , Intestines/physiology , Rotavirus/metabolism , Animals , Anoctamin-1/metabolism , Biological Transport/physiology , Caco-2 Cells , Calcium/metabolism , Cell Line, Tumor , Chlorides/metabolism , Glycoproteins/metabolism , Humans , Male , Mice , Sodium/metabolism , Sodium-Glucose Transporter 1/metabolism , Toxins, Biological/metabolism , Viral Nonstructural Proteins/metabolism
6.
Sci Rep ; 6: 37220, 2016 11 23.
Article in English | MEDLINE | ID: mdl-27876791

ABSTRACT

Destruction of clonogenic cells in the crypt following irradiation are thought to cause altered gastrointestinal function. Previously, we found that an amino acid-based oral rehydration solution (AA-ORS) improved gastrointestinal function in irradiated mice. However, the exact mechanisms were unknown. Electrophysiology, immunohistochemistry, qPCR, and Western blot analysis were used to determine that AA-ORS increased proliferation, maturation, and differentiation and improved electrolyte and nutrient absorption in irradiated mice. A single-hit, multi-target crypt survival curve showed a significant increase in crypt progenitors in irradiated mice treated with AA-ORS for six days (8.8 ± 0.4) compared to the saline-treated group (6.1 ± 0.3; P < 0.001) without a change in D0 (4.8 ± 0.1 Gy). The Dq values increased from 8.8 ± 0.4 Gy to 10.5 ± 0.5 Gy with AA-ORS treatment (P < 0.01), indicating an increased radiation tolerance of 1.7 Gy. We also found that AA-ORS treatment (1) increased Lgr5+, without altering Bmi1 positive cells; (2) increased levels of proliferation markers (Ki-67, p-Erk, p-Akt and PCNA); (3) decreased apoptosis markers, such as cleaved caspase-3 and Bcl-2; and (4) increased expression and protein levels of NHE3 and SGLT1 in the brush border membrane. This study shows that AA-ORS increased villus height and improved electrolyte and nutrient absorption.


Subject(s)
Amino Acids/pharmacology , Cell Proliferation , Gamma Rays/adverse effects , Intestinal Mucosa/metabolism , Radiation Injuries, Experimental/metabolism , Rehydration Solutions/pharmacology , Amino Acids/chemistry , Animals , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Intestinal Mucosa/pathology , Male , Mice , Radiation Injuries, Experimental/drug therapy , Radiation Injuries, Experimental/pathology , Rehydration Solutions/chemistry , Sodium-Glucose Transporter 1/biosynthesis , Sodium-Hydrogen Exchanger 3/biosynthesis
7.
Bioorg Med Chem ; 24(18): 4263-4271, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27448774

ABSTRACT

In an effort to develop new drug candidates with enhanced anticancer activity, our team synthesized and assessed the cytotoxicity of a series of novel xanthone derivatives with two longer 3,6-disubstituted amine carbonyl methoxy side chains on either benzene ring in selected human cancer cell lines. An MTT assay revealed that a set of compounds with lower IC50 values than the positive control, 5-FU, exhibited greater anticancer effects. The most potent derivative (XD8) exhibited anticancer activity in MDA-MB-231, PC-3, A549, AsPC-1, and HCT116 cells lines with IC50 values of 8.06, 6.18, 4.59, 4.76, and 6.09µM, respectively. Cell cycle analysis and apoptosis activation suggested that the mechanism of action of these derivatives includes cell cycle regulation and apoptosis induction.


Subject(s)
Antineoplastic Agents/pharmacology , Xanthones/pharmacology , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Drug Screening Assays, Antitumor , G1 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Structure-Activity Relationship , Xanthones/chemical synthesis
8.
Radiat Res ; 184(5): 509-17, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26488756

ABSTRACT

Triptolide (TPL) may mitigate radiation-induced late pulmonary side effects through its inhibition of global pro-inflammatory cytokines. In this study, we evaluated the effect of TPL in C57BL/6 mice, the animals were exposed to radiation with vehicle (15 Gy), radiation with TPL (0.25 mg/kg i.v., twice weekly for 1, 2 and 3 months), radiation and celecoxib (CLX) (30 mg/kg) and sham irradiation. Cultured supernatant of irradiated RAW 264.7 and MLE-15 cells and lung lysate in different groups were enzyme-linked immunosorbent assays at 33 h. Respiratory rate, pulmonary compliance and pulmonary density were measured at 5 months in all groups. The groups exposed to radiation with vehicle and radiation with TPL exhibited significant differences in respiratory rate and pulmonary compliance (480 ± 75/min vs. 378 ± 76/min; 0.6 ± 0.1 ml/cm H2O/p kg vs. 0.9 ± 0.2 ml/cm H2O/p kg). Seventeen cytokines were significantly reduced in the lung lysate of the radiation exposure with TPL group at 5 months compared to that of the radiation with vehicle group, including profibrotic cytokines implicated in pulmonary fibrosis, such as IL-1ß, TGF- ß1 and IL-13. The radiation exposure with TPL mice exhibited a 41% reduction of pulmonary density and a 25% reduction of hydroxyproline in the lung, compared to that of radiation with vehicle mice. The trichrome-stained area of fibrotic foci and pathological scaling in sections of the mice treated with radiation and TPL mice were significantly less than those of the radiation with vehicle-treated group. In addition, the radiation with TPL-treated mice exhibited a trend of improved survival rate compared to that of the radiation with vehicle-treated mice at 5 months (83% vs. 53%). Three radiation-induced profibrotic cytokines in the radiation with vehicle-treated group were significantly reduced by TPL treatment, and this partly contributed to the trend of improved survival rate and pulmonary density and function and the decreased severity of pulmonary fibrosis at 5 months. Our findings indicate that TPL could be a potential new agent to mitigate radiation-induced pulmonary fibrosis.


Subject(s)
Diterpenes/pharmacology , Phenanthrenes/pharmacology , Pulmonary Fibrosis/drug therapy , Radiation Pneumonitis/drug therapy , Radiation-Protective Agents/pharmacology , Animals , Collagen/metabolism , Cytokines/biosynthesis , Diterpenes/therapeutic use , Epoxy Compounds/pharmacology , Epoxy Compounds/therapeutic use , Female , Lung/drug effects , Lung/pathology , Lung/physiopathology , Lung/radiation effects , Mice , Mice, Inbred C57BL , Organ Size/drug effects , Organ Size/radiation effects , Phenanthrenes/therapeutic use , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/physiopathology , RAW 264.7 Cells , Radiation Pneumonitis/metabolism , Radiation Pneumonitis/pathology , Radiation Pneumonitis/physiopathology , Radiation-Protective Agents/therapeutic use , Survival Rate
9.
Radiat Res ; 183(2): 188-95, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25574588

ABSTRACT

We developed a simple, rapid and quantitative assay using the fluorescent probe PicoGreen to measure the concentration of ionizing radiation-induced double-stranded DNA (dsDNA) in mouse plasma, and we correlated this concentration with the radiation dose. With 70 µl of blood obtained by fingerstick, this 30 min assay reduces protein interference without extending sample processing time. Plasma from nonirradiated mice (BALB/c and NIH Swiss) was pooled, diluted and spiked with dsDNA to establish sensitivity and reproducibility of the assay to quantify plasma dsDNA. The assay was then used to directly quantify dsDNA in plasma at 0-48 h after mice received 0-10 Gy total-body irradiation (TBI). There are three optimal conditions for this assay: 1:10 dilution of plasma in water; 1:200 dilution of PicoGreen reagent in water; and calibration of radiation-induced dsDNA concentration through a standard addition method using serial spiking of samples with genomic dsDNA. Using the internal standard calibration curve of the spiked samples method, the signal developed within 5 min, exhibiting a linear signal (r(2) = 0.997). The radiation-induced elevation of plasma DNA in mice started at 1-3 h, peaked at 9 h and gradually returned to baseline at 24 h after TBI (6 Gy). DNA levels in plasma collected from mice 9 h after 0-10 Gy TBI correlated strongly with dose (r(2) = 0.991 and 0.947 for BALB/c and NIH Swiss, respectively). Using the PicoGreen assay, we observed a radiation dose-dependent response in extracellular plasma DNA 9 h after irradiation with an assay time ≤ 30 min.


Subject(s)
Biological Assay/methods , DNA Damage , DNA, Circular/blood , DNA, Circular/radiation effects , Radiation Monitoring/methods , Animals , DNA, Circular/chemistry , Dose-Response Relationship, Radiation , Fluorescent Dyes/chemistry , Fluorescent Dyes/radiation effects , Male , Mice , Mice, Inbred BALB C , Organic Chemicals/chemistry , Organic Chemicals/radiation effects , Radiation Dosage , Reproducibility of Results , Sensitivity and Specificity , Whole-Body Irradiation
10.
Int J Cell Biol ; 2014: 850460, 2014.
Article in English | MEDLINE | ID: mdl-24688546

ABSTRACT

We investigated whether genetic radiosensitivity-related changes in mtDNA/nDNA ratios are significant to mitochondrial function and if a material effect on mtDNA content and function exists. BALB/c (radiosensitive), C57BL/6 (radioresistant), and F1 hybrid mouse strains were exposed to total body irradiation. Hepatic genomic DNA was extracted, and mitochondria were isolated. Mitochondrial oxygen consumption, ROS, and calcium-induced mitochondrial swelling were measured. Radiation influenced strain-specific survival in vivo. F1 hybrid survival was influenced by maternal input. Changes in mitochondrial content corresponded to survival in vivo among the 4 strains. Calcium-induced mitochondrial swelling was strain dependent. Isolated mitochondria from BALB/c mice were significantly more sensitive to calcium overload than mitochondria from C57BL/6 mice. Maternal input partially influenced the recovery effect of radiation on calcium-induced mitochondrial swelling in F1 hybrids; the hybrid with a radiosensitive maternal lineage exhibited a lower rate of recovery. Hybrids had a survival rate that was biased toward maternal input. mtDNA content and mitochondrial permeability transition pores (MPTP) measured in these strains before irradiation reflected a dominant input from the parent. After irradiation, the MPTP opened sooner in radiosensitive and hybrid strains, likely triggering intrinsic apoptotic pathways. These findings have important implications for translation into predictors of radiation sensitivity/resistance.

11.
Health Phys ; 106(6): 734-44, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24776907

ABSTRACT

Electrolyte and nutrient absorption occur in villous epithelial cells. Radiation often results in reduced electrolyte and nutrient absorption, which leads to gastrointestinal toxicity. Therefore, the authors studied: (1) radiation-induced changes in glucose and amino acid absorption across ileal tissues and (2) the effect of amino acid mixtures on absorptive capacity. NIH Swiss mice were irradiated (0, 1, 3, 5, or 7 Gy) using a ¹³7Cs source at 0.9 Gy min⁻¹. Transepithelial short circuit current (I(sc)), dilution potential, and isotope flux determinations were made in Ussing chamber studies and correlated to plasma endotoxin and IL-1ß levels. Amino acids that increased electrolyte absorption and improved mucosal barrier functions were used to create a mitigating amino acid mixture (MAAM). The MAAM was given to mice via gastric gavage; thereafter, body weight and survival were recorded. A significant decrease in basal and glucose-stimulated sodium absorption occurred after 0, 1, 3, 5, and 7 Gy irradiation. Ussing chamber studies showed that paracellular permeability increased following irradiation and that the addition of glucose resulted in a further increase in permeability. Following irradiation, certain amino acids manifested decreased absorption, whereas others were associated with increased absorption. Lysine, aspartic acid, glycine, isoleucine, threonine, tyrosine, valine, tryptophan, and serine decreased plasma endotoxins were selected for the MAAM. Mice treated with the MAAM showed increased electrolyte absorption and decreased paracellular permeability, IL-1ß levels, and plasma endotoxin levels. Mice treated with MAAM also had increased weight gain and better survival following irradiation. The MAAM has immediate potential for use in mitigating radiation-induced acute gastrointestinal syndrome.


Subject(s)
Amino Acids/chemistry , Amino Acids/pharmacology , Intestines/drug effects , Intestines/radiation effects , Radiation Injuries, Experimental/prevention & control , Radiation-Protective Agents/chemistry , Radiation-Protective Agents/pharmacology , Animals , Dose-Response Relationship, Radiation , Endotoxins/blood , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Glucose/pharmacology , Interleukin-1beta/blood , Intestinal Absorption/drug effects , Intestinal Absorption/radiation effects , Intestinal Mucosa/metabolism , Male , Mice , Permeability/drug effects , Permeability/radiation effects , Radiation Injuries, Experimental/blood , Radiation Injuries, Experimental/metabolism , Sodium/metabolism , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/metabolism , Weight Loss/drug effects , Weight Loss/radiation effects
12.
Adv Exp Med Biol ; 812: 149-155, 2014.
Article in English | MEDLINE | ID: mdl-24729227

ABSTRACT

The tumor vascular system, which is critical to the survival and growth of solid tumors, has been an attractive target for anticancer research. Building on studies that show that some flavonoids have anticancer vascular effects, we developed and analyzed the flavonoid derivative R24 [3, 6-bis (2-oxiranylmethoxy)-9H-xanthen-9-one]. A CAM assay revealed that R24 disrupted neovascular formation; fewer dendrites were detected and overall dendritic length was shorter in the R24-treated chicken embryos. The antiproliferative effect of R24 was measured by MTT assay in A549 (lung cancer), AsPC-1 (pancreatic cancer), HCT-116 (colorectal cancer), and PC-3 (prostate cancer) cell lines. R24 reduced proliferation with an IC50 of 3.44, 3.59, 1.22, and 11.83 µM, respectively. Cell-cycle analysis and Annexin-V/propidium iodide staining showed that R24 induced apoptosis. In addition, R24 regulated intracellular ROS production in a dose-dependent manner. CM-H2DCFDA staining indicated that intracellular ROS production increased with the R24 dose. In summary, we found that R24 exhibits potent antiangiogenic and antiproliferative effects, induces apoptosis, and promotes ROS production.


Subject(s)
Flavonoids/pharmacology , Neoplasms/blood supply , Neovascularization, Pathologic/prevention & control , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Humans
13.
Am J Physiol Cell Physiol ; 306(7): C687-96, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24477233

ABSTRACT

The sodium-coupled glucose transporter-1 (SGLT1)-based oral rehydration solution (ORS) used in the management of acute diarrhea does not substantially reduce stool output, despite the fact that glucose stimulates the absorption of sodium and water. To explain this phenomenon, we investigated the possibility that glucose might also stimulate anion secretion. Transepithelial electrical measurements and isotope flux measurements in Ussing chambers were used to study the effect of glucose on active chloride and fluid secretion in mouse small intestinal cells and human Caco-2 cells. Confocal fluorescence laser microscopy and immunohistochemistry measured intracellular changes in calcium, sodium-glucose linked transporter, and calcium-activated chloride channel (anoctamin 1) expression. In addition to enhancing active sodium absorption, glucose increased intracellular calcium and stimulated electrogenic chloride secretion. Calcium imaging studies showed increased intracellular calcium when intestinal cells were exposed to glucose. Niflumic acid, but not glibenclamide, inhibited glucose-stimulated chloride secretion in mouse small intestines and in Caco-2 cells. Glucose-stimulated chloride secretion was not seen in ileal tissues incubated with the intracellular calcium chelater BAPTA-AM and the sodium-potassium-2 chloride cotransporter 1 (NKCC1) blocker bumetanide. These observations establish that glucose not only stimulates active Na absorption, a well-established phenomenon, but also induces a Ca-activated chloride secretion. This may explain the failure of glucose-based ORS to markedly reduce stool output in acute diarrhea. These results have immediate potential to improve the treatment outcomes for acute and/or chronic diarrheal diseases by replacing glucose with compounds that do not stimulate chloride secretion.


Subject(s)
Chloride Channels/metabolism , Chlorides/metabolism , Glucose/metabolism , Ileum/metabolism , Intestinal Mucosa/metabolism , Animals , Anoctamin-1 , Biological Transport , Caco-2 Cells , Calcium/metabolism , Chelating Agents/pharmacology , Chloride Channels/drug effects , Electric Impedance , Humans , Ileum/drug effects , Immunohistochemistry , Intestinal Mucosa/drug effects , Kinetics , Male , Membrane Transport Modulators/pharmacology , Mice , Microscopy, Confocal , Neoplasm Proteins/metabolism , Sodium/metabolism , Sodium-Glucose Transporter 1/metabolism
14.
Radiat Res ; 180(4): 407-13, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24059680

ABSTRACT

This study demonstrates that mice, similar to humans, have a common mitochondrial DNA deletion (3,860 bp) that encodes 5 transfer RNA genes and 5 polypeptide genes that is related to aging, tissue type and radiotoxicity. Our research indicates that the deletion ratio in the liver was significantly higher than in the brain and gut tissues of 8-month-old mice, as compared to 8-week-old mice. Our results also demonstrate that tissue type, oxidative metabolic capacity and radiosensitivity influence the 3,860-bp deletion level. Therefore, this 3,860-bp deletion content may serve as a biomarker of aging and oxidative damage in mice.


Subject(s)
DNA, Mitochondrial/genetics , Gene Deletion , Mitochondria/genetics , Mitochondria/radiation effects , Aging/genetics , Aging/radiation effects , Animals , Male , Mice , Mice, Inbred C57BL , RNA, Transfer/genetics
15.
Adv Exp Med Biol ; 789: 195-201, 2013.
Article in English | MEDLINE | ID: mdl-23852495

ABSTRACT

Amifostine is a first-line cytoprotective drug used to prevent radiotherapy-induced or chemotherapy-induced injuries. However, its mechanism of action is not well understood. In this study, freshly harvested bone marrow cells were treated with amifostine and analyzed with a series of mitochondrial indices. In vitro results showed that bone marrow cells treated with amifostine 0.5 h before irradiation (0.5 Gy) experienced several benefits, as compared to vehicle controls, including (1) reduced reactive oxygen species levels, which reduced the production of free radicals; (2) better preservation of mitochondria, as indicated by MitoTracker-positive staining and the increased intensity of staining; (3) reduced apoptosis, as demonstrated by Annexin V staining; and (4) a better proliferation rate, as illustrated by MTT assay. Our in vitro studies showed that amifostine-treated mice exhibited (1) higher ATP production; (2) reduced plasma IL-2 levels, suppressing the immune response triggered by radiotoxicity; and (3) enhanced radiation-induced production of granulocyte colony-stimulating factor. All of these processes benefit recovery from radiation-induced damage.


Subject(s)
Amifostine/pharmacology , Bone Marrow Cells/drug effects , Bone Marrow/drug effects , Cytokines/metabolism , Mitochondria/drug effects , Radiation-Protective Agents/pharmacology , Adaptation, Physiological/drug effects , Adenosine Triphosphate/metabolism , Animals , Bone Marrow/growth & development , Bone Marrow/metabolism , Bone Marrow Cells/metabolism , Male , Mice , Mitochondria/metabolism , Reactive Oxygen Species/metabolism
16.
Adv Exp Med Biol ; 789: 257-264, 2013.
Article in English | MEDLINE | ID: mdl-23852503

ABSTRACT

Interleukin 11 (IL-11) is a multifunctional cytokine isolated from bone marrow (BM)-derived stromal cells that promotes hematopoiesis and prolongs the life span of lethally irradiated animals. However, the underlying mechanism for the protective effect of IL-11 on BM is unclear. In this study, we explored the effect of IL-11 on irradiated BM cells. Freshly harvested BM cells were pretreated with 20 ng/ml of recombinant IL-11 for 30 min, irradiated with a dose of 0.5 Gy, cultured for 24 h, and then subjected to several assays. In vitro data showed that, as compared to the vehicle controls, IL-11: (1) reduced the production of reactive oxygen species; (2) reduced the alteration of mitochondrial membrane potential; (3) increased MitoTracker staining, suggesting that the number of mitochondria and their functions were better maintained; and (4) reduced apoptosis of BM cells and enhanced BM cell proliferation. In vivo studies of mice pretreated with saline or 100 µg/kg of IL-11 at 12 and 2 h before 10-Gy total body irradiation (TBI) demonstrated that G-CSF and IL-6 were significantly upregulated, whereas IL-2 and IL-4 were reduced. We found that IL-11 protects mitochondrial functions, acts with G-CSF and IL-6 to stimulate the growth of radiation-damaged BM, and reduces the immune response to radiation injury.


Subject(s)
Bone Marrow/drug effects , Bone Marrow/radiation effects , Interleukin-11/pharmacology , Mitochondria/drug effects , Mitochondria/radiation effects , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/prevention & control , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Bone Marrow/metabolism , Bone Marrow Cells/metabolism , Bone Marrow Cells/radiation effects , Cell Growth Processes/drug effects , Cell Growth Processes/radiation effects , Granulocyte Colony-Stimulating Factor/metabolism , Interleukin-11/metabolism , Interleukin-6/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/radiation effects , Mice , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Whole-Body Irradiation/methods
17.
Adv Exp Med Biol ; 789: 273-280, 2013.
Article in English | MEDLINE | ID: mdl-23852505

ABSTRACT

In this study, we investigated the response of irradiated bone marrow cells to granulocyte colony-stimulating factor (G-CSF). Freshly harvested bone marrow cells were treated with either saline (vehicle control) or 20 ng/ml of G-CSF. Thereafter, cells were separated into nonirradiated (no-IR) and irradiated (IR, 0.5 Gy) groups. IR cells exhibited a higher proliferation rate in response to G-CSF, as compared to the no-IR cells. Reduced levels of reactive oxygen species indicated that G-CSF-treated IR cells produced fewer free radicals, as compared to the no-IR cells. The G-CSF-treated IR cells also had a lower apoptotic rate than their no-IR counterparts. Furthermore, G-CSF-treated IR cells exhibited less alteration of mitochondrial membrane potential, as compared to the no-IR cells. Finally, the mitochondrial number increased in the G-CSF-treated IR cells. The radiation-induced increase in plasma IL-6 in vivo could be enhanced by the administration of G-CSF. The data suggest that radiation potentiates the response of bone marrow cells to G-CSF treatment.


Subject(s)
Bone Marrow/drug effects , Bone Marrow/radiation effects , Granulocyte Colony-Stimulating Factor/pharmacology , Animals , Bone Marrow/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/radiation effects , Free Radicals/metabolism , Interleukin-6/blood , Male , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/radiation effects , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/radiation effects , Reactive Oxygen Species/metabolism
18.
Adv Exp Med Biol ; 765: 335-341, 2013.
Article in English | MEDLINE | ID: mdl-22879053

ABSTRACT

Inflammatory molecules (IMs) play an important role in ionizing radiation (IR)-induced soft tissue damage. The alteration of IMs as a function of time was studied with a protein array containing 62 IMs in mouse cutaneous soft tissues exposed to 30 Gy. The results showed that: (1) 2 days after irradiation, the levels of TGF-ß1, MIP-1γ, IL-1α, and sTNF RI increased, while IGFBP-3, CXCL16, and IL-1ß decreased in IR skin as compared to control skin; (2) 21 days after IR, TGF-ß1, and MIP-1 γ, IL-1α remained high, while CXCL16 and IL-1ß remained low; (3) 3 months after IR, the cytokine pattern exhibited reversals. The levels of MIP-1γ decreased, while VCAM-1, IGFBP-3, and TGF-ß1 production increased. The data indicated that: (a) IMs change as a function of time after soft tissue irradiation; (b) changing IM levels may reflect the altered balance of the cytokine network, leading to imbalance or homeostasis; and (c) an antibody-based protein array can be used to assess multiple IMs simultaneously, making it useful for bulk screening for changes in tissue cytokine levels.


Subject(s)
Hindlimb/metabolism , Hindlimb/radiation effects , Inflammation Mediators/metabolism , Skin/metabolism , Skin/radiation effects , Soft Tissue Injuries/metabolism , Soft Tissue Injuries/pathology , Animals , Chemokines/metabolism , Cytokines/metabolism , Female , Hindlimb/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Protein Array Analysis , Skin/immunology , Time Factors
19.
Adv Exp Med Biol ; 765: 371-377, 2013.
Article in English | MEDLINE | ID: mdl-22879058

ABSTRACT

Mitochondrial DNA (mtDNA) is maternally inherited and controls the oxygen-related production of adenosine-5'-triphosphate, which is transported from the mitochondria to other cellular compartments and used as energy for cellular activities. The mtDNA is physically separated from nuclear DNA (nDNA). Ionizing radiation (IR) causes the release of both mtDNA and nDNA into circulation. Our previous study demonstrated that nDNA has potential to be a biodosimeter. In this study, branched DNA technology was used to explore the alteration pattern of mtDNA after IR. C57BL/6 mice were exposed to 0, 1.5, 3, 6, 8, or 10 Gy total body irradiation; thereafter, plasma mtDNA was assessed with samples collected at 3, 6, 9, 15, 24, 48, 72, or 168 h. We found that: (1) the designed probesets were specific for mtDNA extracted from the liver, and they recognized the small amount of mtDNA mixed in the nDNA; (2) plasma mtDNA exhibited a statistically significant increase only at 6 h after 8 Gy irradiation. The alteration of mtDNA was not dose-dependent or time-dependent; hence, it is unlikely to be an effective biodosimeter.


Subject(s)
Cell Nucleus/genetics , Cell Nucleus/radiation effects , DNA, Mitochondrial/blood , DNA, Mitochondrial/genetics , Radiation Injuries/diagnosis , Whole-Body Irradiation , Animals , Male , Mice , Mice, Inbred C57BL , Radiation Dosage , Radiation Injuries/blood , Radiation Injuries/genetics , Time Factors
20.
PLoS One ; 7(10): e46560, 2012.
Article in English | MEDLINE | ID: mdl-23082125

ABSTRACT

Little is known about the effects of ionizing radiation on the transition and the related signal transduction of progenitor B cells in the bone marrow. Thus, using an NIH Swiss mouse model, we explored the impact of ionizing radiation on the early stage of B-cell development via an examination of the transition of CLP to pro-B to pre-B cells within bone marrow as a function of radiation doses and times. Our results showed that while the total number of bone marrow lymphoid cells at different stages were greatly reduced by subtotal body irradiation (sub-TBI), the surviving cells continued to transition from common lymphoid progenitors to pro-B and then to pre-B in a reproducible temporal pattern. The rearrangement of the immunoglobulin heavy chain increased significantly 1-2 weeks after irradiation, but no change occurred after 3-4 weeks. The rearrangement of the immunoglobulin light chain decreased significantly 1-2 weeks after sub-TBI but increased dramatically after 3-4 weeks. In addition, several key transcription factors and signaling pathways were involved in B-precursor transitions after sub-TBI. The data indicate that week 2 after irradiation is a critical time for the transition from pro-B cells to pre-B cells, reflecting that the functional processes for different B-cell stages are well preserved even after high-dose irradiation.


Subject(s)
Bone Marrow/physiology , Bone Marrow/radiation effects , Precursor Cells, B-Lymphoid/metabolism , Precursor Cells, B-Lymphoid/radiation effects , Regeneration/radiation effects , Whole-Body Irradiation , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Marrow Cells/radiation effects , Cell Count , Gene Rearrangement, B-Lymphocyte , Male , Mice , Precursor Cells, B-Lymphoid/cytology , Receptors, Interleukin-7/metabolism , Signal Transduction/radiation effects , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...