Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 12(9): e0185272, 2017.
Article in English | MEDLINE | ID: mdl-28957383

ABSTRACT

The effect of gestational hypoxia on the neonatal leptin surge, development of hypothalamic arcuate nuclei (ARH) projections and appetite that could contribute to the programming of offspring obesity is lacking. We examined the effect of 12% O2 from gestational days 15-19 in the Sprague-Dawley rat on post-weaning appetite, fat deposition by MRI, adipose tissue cytokine expression, the neonatal leptin surge, ARH response to exogenous leptin, and αMSH projections to the paraventricular nucleus (PVN) in response to a high fat (HFD) or control diet (CD) in male offspring. Normoxia (NMX) and Hypoxia (HPX) offspring exhibited increased food intake when fed a HFD from 5-8 weeks post-birth; HPX offspring on the CD had increased food intake from weeks 5-7 vs. NMX offspring on a CD. HPX offspring on a HFD remained hyperphagic through 23 weeks. Body weight were the same between offspring from HPX vs. NMX dams from 4-12 weeks of age fed a CD or HFD. By 14-23 weeks of age, HPX offspring fed the CD or HFD as well as male NMX offspring fed the HFD were heavier vs. NMX offspring fed the CD. HPX offspring fed a CD exhibited increased abdominal adiposity (MRI) that was amplified by a HFD. HPX offspring fed a HFD exhibited the highest abdominal fat cytokine expression. HPX male offspring had higher plasma leptin from postnatal day (PN) 6 through 14 vs. NMX pups. HPX offspring exhibited increased basal c-Fos labeled cells in the ARH vs. NMX pups on PN16. Leptin increased c-Fos staining in the ARH in NMX but not HPX offspring at PN16. HPX offspring had fewer αMSH fibers in the PVN vs. NMX offspring on PN16. In conclusion, gestational hypoxia impacts the developing ARH resulting in hyperphagia contributing to adult obesity on a control diet and exacerbated by a HFD.


Subject(s)
Hyperphagia/blood , Hyperphagia/complications , Hypoxia/blood , Hypoxia/complications , Leptin/blood , Obesity/blood , Obesity/complications , Adipose Tissue/metabolism , Animals , Animals, Newborn , Anxiety/blood , Anxiety/complications , Arcuate Nucleus of Hypothalamus/metabolism , Behavior, Animal , Diet , Fear , Feeding Behavior , Female , Fetal Weight , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Magnetic Resonance Imaging , Male , Maternal Nutritional Physiological Phenomena , Maze Learning , Motor Activity , Pregnancy , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Water , Weaning , alpha-MSH/metabolism
2.
Neuro Oncol ; 19(2): 175-185, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27416955

ABSTRACT

Background: Despite current therapies, glioblastoma is a devastating cancer, and validation of effective biomarkers for it will enable better diagnosis and therapeutic intervention for this disease. We recently discovered a new biomarker for high-grade gliomas, ELTD1 (epidermal growth factor, latrophilin, and 7 transmembrane domain-containing protein 1 on chromosome 1) via bioinformatics, and validated that ELTD1 protein levels are significantly higher in human and rodent gliomas. The focus of this study was to assess the effect on tumor growth of an antibody against ELTD1 in orthotopic, GL261, and G55 xenograft glioma models. Methods: The effect of anti-ELTD1 antibody therapy was assessed by animal survival, MRI measured tumor volumes, MR angiography, MR perfusion imaging, and immunohistochemistry (IHC) characterization of microvessel density in mouse glioma models. Comparative treatments included anti-vascular endothelial growth factor (VEGF) and anti-c-Met antibody therapies, compared with untreated controls. Results: Tumor volume and survival data in this study show that antibodies against ELTD1 inhibit glioma growth just as effectively or even more so compared with other therapeutic targets studied, including anti-VEGF antibody therapy. Untreated GL261 or G55 tumors were found to have significantly higher ELTD1 levels (IHC) compared with contralateral normal brain. The anti-angiogenic effect of ELTD1 antibody therapy was observed in assessment of microvessel density, as well as from MR angiography and perfusion measurements, which indicated that anti-ELTD1 antibody therapy significantly decreased vascularization compared with untreated controls. Conclusions: Either as a single therapy or in conjunction with other therapeutic approaches, anti-ELTD1 antibodies could be a valuable new clinical anti-angiogenic therapeutic for high-grade gliomas.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal/pharmacology , Brain Neoplasms/drug therapy , Glioma/drug therapy , Neovascularization, Pathologic/prevention & control , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Apoptosis/drug effects , Brain Neoplasms/blood supply , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Disease Models, Animal , Glioma/blood supply , Glioma/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
J Clin Invest ; 125(12): 4349-64, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26571402

ABSTRACT

Tumor angiogenesis is critical for cancer progression. In multiple murine models, endothelium-specific epsin deficiency abrogates tumor progression by shifting the balance of VEGFR2 signaling toward uncontrolled tumor angiogenesis, resulting in dysfunctional tumor vasculature. Here, we designed a tumor endothelium-targeting chimeric peptide (UPI) for the purpose of inhibiting endogenous tumor endothelial epsins by competitively binding activated VEGFR2. We determined that the UPI peptide specifically targets tumor endothelial VEGFR2 through an unconventional binding mechanism that is driven by unique residues present only in the epsin ubiquitin-interacting motif (UIM) and the VEGFR2 kinase domain. In murine models of neoangiogenesis, UPI peptide increased VEGF-driven angiogenesis and neovascularization but spared quiescent vascular beds. Further, in tumor-bearing mice, UPI peptide markedly impaired functional tumor angiogenesis, tumor growth, and metastasis, resulting in a notable increase in survival. Coadministration of UPI peptide with cytotoxic chemotherapeutics further sustained tumor inhibition. Equipped with localized tumor endothelium-specific targeting, our UPI peptide provides potential for an effective and alternative cancer therapy.


Subject(s)
Adaptor Proteins, Vesicular Transport/pharmacology , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Peptides/pharmacology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Amino Acid Motifs , Animals , Mice , Mice, Knockout , Neoplasm Metastasis , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Peptides/genetics , Peptides/metabolism , Protein Structure, Tertiary , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
5.
Am J Nucl Med Mol Imaging ; 5(4): 363-78, 2015.
Article in English | MEDLINE | ID: mdl-26269774

ABSTRACT

Angiogenesis is essential to tumor progression, and the precise imaging of the angiogenic marker vascular endothelial growth factor receptor 2 (VEGFR-2) may provide an accurate evaluation for angiogenesis during a therapeutic response. With the use of molecular magnetic resonance imaging (mMRI), an in vitro cell assay indicated significantly decreased T1 relaxation values when tumor endothelial cells (TEC), which positively expressed VEGFR-2 (Western blot), were in the presence of the VEGFR-2 probe compared to TEC alone (P < 0.001). For in vivo mMRI evaluations, we assessed VEGFR-2 levels in untreated and OKN-007-treated GL261 mouse gliomas. Regarding treatment response, OKN-007 was also able to significantly decrease tumor volumes (P < 0.01) and increase survival (P < 0.001) in treated animals. Regarding in vivo detection of VEGFR-2, OKN-007 was found to significantly decrease the amount of VEGFR-2 probe (P < 0.05) compared to an untreated control group. Fluorescence imaging for the VEGFR-2 probe indicated that there was colocalization with the endothelial marker CD31 in an untreated tumor bearing mouse and decreased levels for an OKN-007-treated animal. Immuno-fluorescence imaging for VEGFR-2 indicated that OKN-007 treatment significantly decreased VEGFR-2 levels (P < 0.0001) when compared to untreated tumors. Immuno-electron microscopy was used with gold-labeled anti-biotin to detect the anti-VEGFR-2 probe within the plasma membrane of GL261 tumor endothelial cells. This is the first attempt at detecting in vivo levels of VEGFR-2 in a mouse GL261 glioma model and assessing the anti-angiogenic capability of an anticancer nitrone. The results indicate that OKN-007 treatment substantially decreased VEGFR-2 levels in a GL261 glioma model, and can be considered as an anti-angiogenic therapy in human gliomas.

6.
Free Radic Biol Med ; 87: 157-68, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26119786

ABSTRACT

Free radicals are associated with glioma tumors. Here, we report on the ability of an anticancer nitrone compound, OKN-007 [Oklahoma Nitrone 007; a disulfonyl derivative of α-phenyl-tert-butyl nitrone (PBN)] to decrease free radical levels in F98 rat gliomas using combined molecular magnetic resonance imaging (mMRI) and immunospin-trapping (IST) methodologies. Free radicals are trapped with the spin-trapping agent, 5,5-dimethyl-1-pyrroline N-oxide (DMPO), to form DMPO macromolecule radical adducts, and then further tagged by immunospin trapping by an antibody against DMPO adducts. In this study, we combined mMRI with a biotin-Gd-DTPA-albumin-based contrast agent for signal detection with the specificity of an antibody for DMPO nitrone adducts (anti-DMPO probe), to detect in vivo free radicals in OKN-007-treated rat F98 gliomas. OKN-007 was found to significantly decrease (P < 0.05) free radical levels detected with an anti-DMPO probe in treated animals compared to untreated rats. Immunoelectron microscopy was used with gold-labeled antibiotin to detect the anti-DMPO probe within the plasma membrane of F98 tumor cells from rats administered anti-DMPO in vivo. OKN-007 was also found to decrease nuclear factor erythroid 2-related factor 2, inducible nitric oxide synthase, 3-nitrotyrosine, and malondialdehyde in ex vivo F98 glioma tissues via immunohistochemistry, as well as decrease 3-nitrotyrosine and malondialdehyde adducts in vitro in F98 cells via ELISA. The results indicate that OKN-007 effectively decreases free radicals associated with glioma tumor growth. Furthermore, this method can potentially be applied toward other types of cancers for the in vivo detection of macromolecular free radicals and the assessment of antioxidants.


Subject(s)
Antioxidants/administration & dosage , Benzenesulfonates/administration & dosage , Free Radicals/metabolism , Glioma/drug therapy , Imines/administration & dosage , Animals , Contrast Media/chemistry , Cyclic N-Oxides/chemistry , Disease Models, Animal , Free Radicals/chemistry , Glioma/metabolism , Glioma/pathology , Humans , Magnetic Resonance Imaging , Male , Malondialdehyde/chemistry , Malondialdehyde/metabolism , Rats , Spin Trapping
7.
J Magn Reson Imaging ; 42(6): 1582-91, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25920494

ABSTRACT

BACKGROUND: Glioblastoma is a malignant World Health Organization (WHO) grade IV glioma with a poor prognosis in humans. New therapeutics are desperately required. The nitrone OKN-007 (2,4-disulfophenyl-PBN) has demonstrated effective anti-glioma properties in several rodent models and is currently being used as a clinical investigational drug for recurrent gliomas. We assessed the regional effects of OKN-007 in the tumor necrotic core and non-necrotic tumor parenchyma. METHODS: An F98 rat glioma model was evaluated using proton magnetic resonance spectroscopy ((1) H-MRS), diffusion-weighted imaging (DWI), morphological T2-weighted imaging (T2W) at 7 Tesla (30 cm-bore MRI), as well as immunohistochemistry and microarray assessments, at maximum tumor volumes (15-23 days following cell implantation in untreated (UT) tumors, and 18-35 days in OKN-007-treated tumors). RESULTS: (1) H-MRS data indicates that Lip0.9/Cho, Lip0.9/Cr, Lip1.3/Cho, and Lip1.3/Cr ratios are significantly decreased (all P < 0.05) in the OKN-007-treated group compared with UT F98 gliomas. The Cho/Cr ratio is also significantly decreased in the OKN-007-treated group compared with UT gliomas. In addition, the OKN-007-treated group demonstrates significantly lower ADC values in the necrotic tumor core and the nonnecrotic tumor parenchyma (both P < 0.05) compared with the UT group. There was also an increase in apoptosis following OKN-007 treatment (P < 0.01) compared with UT. CONCLUSION: OKN-007 reduces both necrosis and tumor cell proliferation, as well as seems to mediate multiple effects in different tumor regions (tumor necrotic core and nonnecrotic tumor parenchyma) in F98 gliomas, indicating the efficacy of OKN-007 as an anti-cancer agent and its potential clinical use.


Subject(s)
Benzenesulfonates/administration & dosage , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Glioma/drug therapy , Glioma/pathology , Imines/administration & dosage , Magnetic Resonance Imaging/methods , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Necrosis/pathology , Necrosis/prevention & control , Rats , Rats, Inbred F344
8.
Neuro Oncol ; 15(3): 330-40, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23328810

ABSTRACT

BACKGROUND: Glioblastoma multiforme, a World Health Organization grade IV glioma, has a poor prognosis in humans despite current treatment options. Here, we present magnetic resonance imaging (MRI) data regarding the regression of aggressive rat F98 gliomas and human U87 glioma xenografts after treatment with the nitrone compound OKN-007, a disulfonyl derivative of α-phenyl-tert-butyl nitrone. METHODS: MRI was used to assess tumor volumes in F98 and U87 gliomas, and bioluminescence imaging was used to measure tumor volumes in F98 gliomas encoded with the luciferase gene (F98(luc)). Immunohistochemistry was used to assess angiogenesis (vascular endothelial growth factor [VEGF] and microvessel density [MVD]), cell differentiation (carbonic anhydrase IX [CA-IX]), hypoxia (hypoxia-inducible factor-1α [HIF-1α]), cell proliferation (glucose transporter 1 [Glut-1] and MIB-1), proliferation index, and apoptosis (cleaved caspase 3) markers in F98 gliomas. VEGF, CA-IX, Glut-1, HIF-1α, and cleaved caspase 3 were assessed in U87 gliomas. RESULTS: Animal survival was found to be significantly increased (P < .001 for F98, P < .01 for U87) in the group that received OKN-007 treatment compared with the untreated groups. After MRI detection of F98 gliomas, OKN-007, administered orally, was found to decrease tumor growth (P < .05). U87 glioma volumes were found to significantly decrease (P < .05) after OKN-007 treatment, compared with untreated animals. OKN-007 administration resulted in significant decreases in tumor hypoxia (HIF-1α [P < .05] in both F98 and U87), angiogenesis (MVD [P < .05], but not VEGF, in F98 or U87), and cell proliferation (Glut-1 [P < .05 in F98, P < .01 in U87] and MIB-1 [P < .01] in F98) and caused a significant increase in apoptosis (cleaved caspase 3 [P < .001 in F98, P < .05 in U87]), compared with untreated animals. CONCLUSIONS: OKN-007 may be considered as a promising therapeutic addition or alternative for the treatment of aggressive human gliomas.


Subject(s)
Apoptosis , Benzenesulfonates/pharmacology , Brain Neoplasms/prevention & control , Cell Proliferation , Glioma/prevention & control , Imines/pharmacology , Neovascularization, Pathologic/prevention & control , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Differentiation , Cell Movement , Glioma/metabolism , Glioma/pathology , Humans , Immunoenzyme Techniques , Luminescent Measurements , Magnetic Resonance Imaging , Rats , Rats, Inbred F344 , Rats, Nude , Survival Rate , Tumor Cells, Cultured
9.
Pancreas ; 41(3): 474-80, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22422139

ABSTRACT

OBJECTIVES: The aims of this study were (1) to determine nuclear magnetic resonance spectroscopic characteristics and metabolite profiles of serum samples from patients with pancreatic cancer compared with noncancerous control samples and (2) to ascertain if the accuracy of metabolite identification by 1D spectra can be improved upon by confirmation of spin-system assignment using more sophisticated experiments. METHODS: Nuclear magnetic resonance spectra, including 1D, total correlation spectroscopy, and heteronuclear multiple/single quantum coherence, were obtained from serum samples from patients with pancreatic cancer and control subjects and used to determine serum levels of a range of metabolites. RESULTS: The data show that total choline (P = 0.03), taurine (P = 0.03), and glucose plus triglycerides (P = 0.01) are significantly higher in cancer versus control samples. Also detected were species that could not be individually identified and that were designated UCM (unresolved complex matter). Levels of UCM are significantly higher in subjects with cancer, being almost double those of control samples. CONCLUSIONS: Although metabolites such as lactate, taurine, glucose, choline, and triglycerides can be determined from 1D spectra, accuracy is improved by confirmation of spin-system assignment with total correlation spectroscopy and heteronuclear multiple/single quantum coherence spectral analysis. In addition, we introduce a new metric, UCM, which is at higher concentrations in cancer compared with control samples.


Subject(s)
Biomarkers, Tumor/blood , Magnetic Resonance Spectroscopy , Metabolomics/methods , Pancreatic Neoplasms/metabolism , Blood Glucose/analysis , Case-Control Studies , Choline/blood , Fatty Acids, Unsaturated/blood , Female , Humans , Lactic Acid/blood , Male , Oklahoma , Pancreatic Neoplasms/blood , Pilot Projects , Taurine/blood , Triglycerides/blood , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...