Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Toxicol Sci ; 148(2): 555-66, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26363170

ABSTRACT

Perinatal environmental exposures are potentially important contributors to the increase in autoimmune diseases. Yet, the mechanisms by which these exposures increase self-reactive immune responses later in life are poorly understood. Autoimmune diseases require CD4(+) T cells for initiation, progression, and/or clinical symptoms; thus, developmental exposures that cause durable changes in CD4(+) T cells may play a role. Early life activation of the aryl hydrocarbon receptor (AHR) causes persistent changes in the response of CD4(+) T cells to infection later in life but whether CD4(+) T cells are affected by developmental exposure in the context of an autoimmune disease is unknown. Gnaq(+/-) mice develop symptoms of autoimmune disease similar to those measured clinically, and therefore can be used to evaluate gene-environment interactions during development on disease progression. Herein, we examined the effect of AHR activation in utero and via lactation, or solely via lactation, on disease onset and severity in adult Gnaq(+/-) offspring. Developmental activation of the AHR-accelerated disease in Gnaq(+/-) mice, and this correlates with increases in effector CD4(+) T-cell populations. Increased symptom onset and cellular changes due to early life AHR activation were more evident in female Gnaq(+/-) mice compared with males. These observations suggest that developmental AHR activation by pollutants, and other exogenous ligands, may increase the likelihood that genetically predisposed individuals will develop clinical symptoms of autoimmune disease later in life.


Subject(s)
Autoimmune Diseases/chemically induced , Autoimmunity , Basic Helix-Loop-Helix Transcription Factors/agonists , CD4-Positive T-Lymphocytes/enzymology , Environmental Pollutants/toxicity , GTP-Binding Protein alpha Subunits, Gq-G11/deficiency , Polychlorinated Dibenzodioxins/toxicity , Receptors, Aryl Hydrocarbon/agonists , Age Factors , Animals , Autoimmune Diseases/enzymology , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Basic Helix-Loop-Helix Transcription Factors/metabolism , CD4-Positive T-Lymphocytes/immunology , Female , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Gene-Environment Interaction , Genetic Predisposition to Disease , Gestational Age , Male , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Pregnancy , Prenatal Exposure Delayed Effects , Receptors, Aryl Hydrocarbon/metabolism , Risk Factors , Severity of Illness Index , Sex Factors
2.
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
3.
Biol Reprod ; 90(5): 110, 2014 May.
Article in English | MEDLINE | ID: mdl-24648400

ABSTRACT

The human placenta performs multiple essential functions required for successful pregnancy. Alterations in the placental vasculature have been implicated in severe complications of pregnancy. Despite the importance of placental vascular function during pregnancy, there are gaps in our knowledge regarding the molecular pathways that control vessel development. Furthermore, there are limited tools available to simultaneously examine the morphology, phenotype, and spatial arrangement of cells within intact placental structures. To overcome these limitations, we developed whole mount immunofluorescence (WMIF) of the human placenta. Morphological analyses using WMIF revealed that blood vessel structures were consistent with an immature, angiogenic morphology in first-trimester placentas and mature, remodeled endothelium at term. To investigate placental expression of factors that control blood vessel development, we utilized WMIF to examine gestation age-specific expression of 1) the receptors for vascular endothelial growth factor (VEGFR-1, VEGFR-2, and VEGFR-3), which are required for placental vascular development in mice, and 2) activated, tyrosine phosphorylated STAT3 (pSTAT3), a transcription factor that mediates VEGFR2 signaling. We detected high levels of VEGFR2, VEGFR3, and pSTAT3 expression in early placental blood vessels that were significantly diminished by term. VEGFR1 was expressed primarily in trophoblast and Hofbauer cells throughout gestation. Based on our collective results, we propose that VEGFR2, VEGFR3, and STAT3 play essential roles in the development of the human placental vasculature. In addition, we anticipate that WMIF will provide a powerful approach for comparing placental morphology and protein expression in normal versus pathological pregnancies and for investigating the effects of environmental factors on placental function.


Subject(s)
Neovascularization, Physiologic/physiology , Placenta/blood supply , Female , Humans , Image Processing, Computer-Assisted , Microscopy, Confocal , Microscopy, Fluorescence , Placenta/metabolism , Placenta/ultrastructure , Pregnancy , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
4.
Int J Radiat Oncol Biol Phys ; 77(1): 261-8, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20394858

ABSTRACT

PURPOSE: Acute gastrointestinal syndrome (AGS) resulting from ionizing radiation causes death within 7 days. Currently, no satisfactory agent exists for mitigation of AGS. A peptide derived from the receptor binding domain of fibroblast growth factor 2 (FGF-P) was synthesized and its mitigation effect on AGS was examined. METHODS AND MATERIALS: A subtotal body irradiation (sub-TBI) model was created to induce gastrointestinal (GI) death while avoiding bone marrow death. After 10.5 to 16 Gy sub-TBI, mice received an intramuscular injection of FGF-P (10 mg/kg/day) or saline (0.2 ml/day) for 5 days; survival (frequency and duration) was measured. Crypt cells and their proliferation were assessed by hematoxylin, eosin, and BrdU staining. In addition, GI hemoccult score, stool formation, and plasma levels of endotoxin, insulin, amylase, interleukin (IL)-6, keratinocyte-derived chemokine (KC) monocyte chemoattractant protein 1 (MCP-1) and tumor necrosis factor (TNF)-alpha were evaluated. RESULTS: Treatment with FGF-P rescued a significant fraction of four strains of mice (33-50%) exposed to a lethal dose of sub-TBI. Use of FGF-P improved crypt survival and repopulation and partially preserved or restored GI function. Furthermore, whereas sub-TBI increased plasma endotoxin levels and several pro-inflammation cytokines (IL-6, KC, MCP-1, and TNF-alpha), FGF-P reduced these adverse responses. CONCLUSIONS: The study data support pursuing FGF-P as a mitigator for AGS.


Subject(s)
Fibroblast Growth Factor 2/therapeutic use , Gastrointestinal Tract/radiation effects , Peptide Fragments/therapeutic use , Radiation Injuries, Experimental/prevention & control , Radiation-Protective Agents/therapeutic use , Animals , Biomarkers/blood , Blood Glucose/drug effects , Bone Marrow/drug effects , Bone Marrow/radiation effects , Chemokine CCL2/blood , Chemokines/blood , Drug Evaluation, Preclinical/methods , Endotoxemia/etiology , Endotoxemia/prevention & control , Gastrointestinal Tract/drug effects , Insulin/blood , Interleukin-6/blood , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Radiation Injuries, Experimental/blood , Radiation Injuries, Experimental/mortality , Species Specificity , Syndrome , Tumor Necrosis Factor-alpha/blood
5.
Mol Med ; 16(1-2): 10-8, 2010.
Article in English | MEDLINE | ID: mdl-19946606

ABSTRACT

The main circulating estrogen hormone 17beta-estradiol (E2) contributes to the initiation and progression of breast cancer. Estrogen receptors (ERs), as transcription factors, mediate the effects of E2. Ablation of the circulating E2 and/or prevention of ER functions constitute approaches for ER-positive breast cancer treatments. These modalities are, however, ineffective in de novo endocrine-resistant breast neoplasms that do not express ERs. The interaction of E2-ERs with specific DNA sequences, estrogen responsive elements (EREs), of genes constitutes one genomic pathway necessary for cellular alterations. We herein tested the prediction that specific regulation of ERE-driven genes by an engineered monomeric and constitutively active transcription factor, monotransregulator, provides a basis for the treatment of ER-negative breast cancer. Using adenovirus infected ER-negative MDA-MB-231 cells derived from a breast adenocarcinoma, we found that the monotransregulator, but not the ERE-binding defective counterpart, repressed cellular proliferation and motility, and induced apoptosis through expression of genes that required ERE interactions. Similarly, the monotransregulator suppressed the growth of ER-negative BT-549 cells derived from a breast-ductal carcinoma. Moreover, the ERE-binding monotransregulator repressed xenograft tumor growth in a nude mice model. Thus, specific regulation of genes bearing EREs could offer a therapeutic approach for de novo endocrine-resistant breast cancers.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/therapy , Estrogen Receptor alpha/genetics , Response Elements , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Death/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Movement/physiology , Estradiol/metabolism , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic , Genes, Regulator , Genetic Therapy/methods , Humans , Mice , Mice, Nude , Xenograft Model Antitumor Assays
6.
Int J Radiat Oncol Biol Phys ; 74(5): 1592-9, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19616745

ABSTRACT

PURPOSE: Current biodosimetric techniques for determining radiation exposure have inherent delays, as well as quantitation and interpretation limitations. We have identified a new technique with the advantage of directly measuring circulating DNA by amplifying inter-B1 regions in the mouse genome, providing a sensitive method for quantitating plasma DNA. METHODS AND MATERIALS: Real-time quantitative polymerase chain reaction (PCR) was used to detect levels of DNA by amplifying inter-B1 genomic DNA in plasma samples collected at 0-48 h from mice receiving 0-10 Gy total- or partial-body irradiation ((137)Cs gamma-ray source at approximately 1.86 Gy/min; homogeneity: +/- 6.5%). RESULTS: The correlation coefficient between DNA levels and the threshold cycle value (C(T)) was 0.996, and the average recoveries of DNA in the assay were 87%. This assay revealed that when BALB/c mice were exposed to 10 Gy total-body irradiation (TBI), plasma DNA levels gradually increased beginning at 3 h after irradiation, peaked at 9 h, and returned to baseline within 48 h. Increased plasma DNA levels were also detected following upper-torso or lower-torso partial-body irradiation; however, TBI approximately doubled those plasma DNA levels at the same radiation dose. This technique therefore reflects total body cell damage. The advantages of this assay are that DNA extraction is not required, the assay is highly sensitive (0.002 ng), and results can be obtained within 2.5 h after collection of plasma samples. CONCLUSIONS: A radiation dose-dependent increase of plasma DNA was observed in the dose range from 2 to 10 Gy, suggesting that plasma DNA may be a useful radiation biomarker and adjunct to existing cell-based assays.


Subject(s)
DNA/blood , Gamma Rays , Polymerase Chain Reaction/methods , Whole-Body Irradiation , Algorithms , Animals , Biomarkers/blood , DNA/radiation effects , DNA Damage , DNA Primers , Dose-Response Relationship, Radiation , Genome/genetics , Genome/radiation effects , Male , Mice , Mice, Inbred BALB C , Time Factors
7.
Radiat Res ; 171(5): 606-14, 2009 May.
Article in English | MEDLINE | ID: mdl-19580496

ABSTRACT

A variety of antiangiogenic strategies have proven effective in preclinical tumor models, either as single agents or in combination with radiation. Clinical gains have been relatively modest, however, and questions remain regarding optimal scheduling. The objectives of the current work were to evaluate whether the sequencing of acute treatment critically affects tumor pathophysiological and therapeutic response. Axitinib (Pfizer Global Research & Development), an inhibitor that predominantly targets vascular endothelial growth factor receptors, was administered either before or after each daily radiation fraction in two human prostate xenograft tumor models. Tumors were frozen at sequential times to monitor changes in (1) vascular spacing, (2) pericyte and basement membrane coverage, and (3) hypoxia. Although similar reductions in blood vessel counts were observed with each tumor model, tumor vasculature was not functionally normalized. Instead, tumor hypoxia increased, accompanied by a progressive dissociation of pericytes and basement membranes. Ultimately, tumor growth inhibition was found to be equivalent for each of the combination schedules. These studies illustrate a clear advantage to combining axitinib with fractionated therapy but argue against an acute radiosensitization or radioprotection of either the tumor cells or tumor vasculature. Instead, post- and preirradiation daily drug administration serve equally well in supplementing the response to radiotherapy.


Subject(s)
Dose Fractionation, Radiation , Imidazoles/pharmacology , Indazoles/pharmacology , Neoplasms/therapy , Protein Kinase Inhibitors/pharmacology , Axitinib , Basement Membrane/drug effects , Basement Membrane/radiation effects , Cell Line, Tumor , Combined Modality Therapy , Humans , Male , Neoplasms/pathology , Pericytes/drug effects , Pericytes/radiation effects
8.
Cancer Res ; 67(20): 9921-8, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17942924

ABSTRACT

Although antiangiogenic strategies have proven highly promising in preclinical studies and some recent clinical trials, generally only combinations with cytotoxic therapies have shown clinical effectiveness. An ongoing question has been whether conventional therapies are enhanced or compromised by antiangiogenic agents. The present studies were designed to determine the pathophysiologic consequences of both single and combined treatments using fractionated radiotherapy plus AG-013736, a receptor tyrosine kinase inhibitor that preferentially inhibits vascular endothelial growth factor receptors. DU145 human prostate xenograft tumors were treated with (a) vehicle alone, (b) AG-013736, (c) 5x2 Gy/wk radiotherapy fractions, or (d) the combination. Automated image processing of immunohistochemical images was used to determine total and perfused blood vessel spacing, overall hypoxia, pericyte/collagen coverage, proliferation, and apoptosis. Combination therapy produced an increased tumor response compared with either monotherapy alone. Vascular density progressively declined in concert with slightly increased alpha-smooth muscle actin-positive pericyte coverage and increased overall tumor hypoxia (compared with controls). Although functional vessel endothelial apoptosis was selectively increased, reductions in total and perfused vessels were generally proportionate, suggesting that functional vasculature was not specifically targeted by combination therapy. These results argue against either an AG-013736- or a combination treatment-induced functional normalization of the tumor vasculature. Vascular ablation was mirrored by the increased appearance of dissociated pericytes and empty type IV collagen sleeves. Despite the progressive decrease in tumor oxygenation over 3 weeks of treatment, combination therapy remained effective and tumor progression was minimal.


Subject(s)
Imidazoles/pharmacology , Indazoles/pharmacology , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/therapy , Actins/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Axitinib , Cell Growth Processes/drug effects , Cell Growth Processes/radiation effects , Cell Hypoxia/drug effects , Cell Hypoxia/radiation effects , Collagen Type IV/metabolism , Combined Modality Therapy , Dose Fractionation, Radiation , Humans , Male , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/radiotherapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor beta/metabolism , Xenograft Model Antitumor Assays
9.
Clin Cancer Res ; 13(16): 4891-9, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17699869

ABSTRACT

PURPOSE: The lack of effective treatment for pancreatic cancer results in a very low survival rate. This study explores the enhancement of the therapeutic effect on human pancreatic cancer via the combination of triptolide and ionizing radiation (IR). EXPERIMENTAL DESIGN: In vitro AsPC-1 human pancreatic cancer cells were treated with triptolide alone, IR alone, or triptolide plus IR. Cell proliferation was analyzed with sulforhodamine B (SRB) method and clonogenic survival; comparison of apoptosis induced by the above treatment was analyzed by annexin V-propidium iodide (PI) staining. Furthermore, the expression of apoptotic pathway intermediates was measured by the assay of caspase activity and Western blot. Mitochondrial transmembrane potential was determined by JC-1 assay. In vivo, AsPC-1 xenografts were treated with 0.25 mg/kg triptolide, 10 Gy IR, or triptolide plus IR. The tumors were measured for volume and weight at the end of the experiment. Tumor tissues were tested for terminal nucleotidyl transferase-mediated nick end labeling (TUNEL) and immunohistochemistry. RESULTS: The combination of triptolide plus IR reduced cell survival to 21% and enhanced apoptosis, compared with single treatment. In vivo, tumor growth of AsPC-1 xenografts was reduced further in the group treated with triptolide plus IR compared with single treatment. TUNEL and immunohistochemistry of caspase-3 cleavage in tumor tissues indicated that the combination of triptolide plus IR resulted in significantly enhanced apoptosis compared with single treatments. CONCLUSIONS: Triptolide in combination with ionizing radiation produced synergistic antitumor effects on pancreatic cancer both in vitro and in vivo and seems promising in the combined modality therapy of pancreatic cancer.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Diterpenes/pharmacology , Pancreatic Neoplasms/therapy , Phenanthrenes/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor , Combined Modality Therapy , Epoxy Compounds/pharmacology , G2 Phase/drug effects , G2 Phase/radiation effects , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Pancreatic Neoplasms/radiotherapy , Transplantation, Heterologous
10.
Med Phys ; 33(6): 1623-33, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16872070

ABSTRACT

Ultrasound-induced blood stasis has been observed for more than 30 years. Most of the literature has been focused on the health risks associated with this phenomenon and methods employed to prevent stasis from occurring during ultrasound imaging. To date, experimental observations have been either in vitro or invasive. The current work demonstrates ultrasound-induced blood stasis in murine normal leg muscle versus tumor-bearing legs, observed through noninvasive measurements of optical spectroscopy, and discusses possible diagnostic uses for this previously undesirable effect of ultrasound. We demonstrate that, using optical spectroscopy, effects of ultrasound can be used to differentiate tumor from normal leg muscle tissue in mice. Finally, we propose a novel diagnostic algorithm that quantitatively differentiates tumor from nontumor with maximum specificity 0.83, maximum sensitivity 0.79, and area under receiver-operating-characteristics curve 0.90.


Subject(s)
Hemostasis/radiation effects , Neoplasms/diagnosis , Optics and Photonics , Spectrum Analysis/methods , Ultrasonics , Animals , Diagnosis, Differential , Disease Models, Animal , Female , Hindlimb/blood supply , Hindlimb/pathology , Hindlimb/radiation effects , Mice , Muscles/blood supply , Muscles/pathology , Muscles/radiation effects , Neoplasms/blood supply , Neoplasms/pathology , Spectrum Analysis/instrumentation , Time Factors
11.
Int J Radiat Oncol Biol Phys ; 65(3): 882-9, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16751070

ABSTRACT

PURPOSE: To investigate the effect of esculentoside A (EsA) on radiation-induced cutaneous and fibrovascular toxicity and its possible molecular mechanisms, both in vivo and in vitro. METHODS AND MATERIALS: Mice received drug intervention 18 hours before 30 Gy to the right hind leg. Alterations in several cytokines expressed in skin tissue 2 days after irradiation were determined by ELISA. Early skin toxicity was evaluated 3 to 4 weeks after irradiation by skin scoring, and both tissue contraction and expression of TGF-beta1 were determined for soft-tissue fibrosis 3 months after irradiation. In vitro, the effect of EsA on radiation-induced nitric oxide (NO) and cytokine production in different cell types was measured by application of 2, 4, and 8 Gy. RESULTS: In vivo, EsA reduced levels of IL-1alpha, MCP-1, VEGF, and TGF-beta1 in cutaneous tissue and reduced soft-tissue toxicity. In vitro, EsA inhibited the IL-1alpha ordinarily produced after 4 Gy in A431 cells. In Raw264.7 cells, EsA reduced levels of IL-1alpha, IL-1beta, and NO production costimulated by radiation and lipopolysaccharide (LPS). In L-929 cells, EsA inhibited VEGF, TNF, and MCP-1 production at 2, 4, and 8 Gy. CONCLUSIONS: Esculentoside A protects soft tissues against radiation toxicity through inhibiting the production of several proinflammatory cytokines and inflammatory mediators in epithelial cells, macrophages, fibroblasts, and skin tissue.


Subject(s)
Drugs, Chinese Herbal/therapeutic use , Oleanolic Acid/analogs & derivatives , Radiodermatitis/prevention & control , Saponins/therapeutic use , Skin/radiation effects , Animals , Cell Line, Tumor/metabolism , Cell Line, Tumor/radiation effects , Cytokines/analysis , Female , Fibroblasts/metabolism , Fibroblasts/radiation effects , Fibrosis/prevention & control , Macrophages/metabolism , Macrophages/radiation effects , Mice , Mice, Inbred C57BL , Nitric Oxide/analysis , Oleanolic Acid/therapeutic use , Radiodermatitis/metabolism , Skin/pathology , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1
12.
Adv Exp Med Biol ; 566: 59-65, 2005.
Article in English | MEDLINE | ID: mdl-16594135

ABSTRACT

Since conventional therapies are directly dependent on the supply of either drugs or oxygen, a key question is whether antiangiogenic agents produce detrimental effects on tumor vascular function, thus compromising combination therapies. A second question is whether experimental results based on fast-growing, transplanted tumors mimic those in slowly developing spontaneous tumors, which may be more representative of response in human primary tumors. To investigate changes in tumor pathophysiology, three antiangiogenic agents were compared: a) endostatin, b) anti-VEGFR-2 (DC101), and c) celecoxib. Total blood vessels were identified using anti-CD31, perfused vessels using DiOC7, and hypoxia by EF5 uptake. Although individual tumor growth rates varied substantially, DC101 produced the most striking inhibition. DC101 increased total and perfused vessel spacing as well as overall hypoxia, while endostatin increased total vessel spacing, and hypoxia and celecoxib had no marked effects. These results reinforce the idea that pathophysiological changes in spontaneous tumors are in general reflective of response in transplanted tumors. Furthermore, although DC101 inhibited growth in roughly half of the spontaneous tumors, the remaining tumors were unaffected. A key focus of future studies will be to investigate the underlying rationale for the widely varying antiangiogenic response among tumors that outwardly appear so similar.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Animals , Celecoxib , Endostatins/pharmacology , Female , Hypoxia/drug therapy , Hypoxia/physiopathology , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/physiopathology , Mice , Mice, Inbred C3H , Pyrazoles/pharmacology , Recombinant Proteins/pharmacology , Sulfonamides/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
13.
Cancer Res ; 64(16): 5712-9, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15313911

ABSTRACT

Although clinical trials of antiangiogenic strategies have been disappointing when administered as single agents, such approaches can play an important role in cancer treatment when combined with conventional therapies. Previous studies have shown that DC101, an antiangiogenic monoclonal antibody against vascular endothelial growth factor receptor-2, can produce significant growth inhibition in spontaneous and transplanted tumors but can also induce substantial hypoxia. Because DC101 appears to potentiate radiotherapy in some tumors, the present studies were undertaken to characterize pathophysiological changes following combined therapy and to determine whether radioresponse is enhanced despite the induction of hypoxia. MCa-4 and MCa-35 mammary carcinomas were treated with: (a) DC101; (b) 5 x 6 Gy radiation fractions; or (c) the combination. Image analysis of frozen tumor sections was used to quantitate: (a) hypoxia; (b) spacing of total and perfused blood vessels; and (c) endothelial and tumor cell apoptosis. For MCa-4, combination treatment schedules produced significant and prolonged delays in tumor growth, whereas single-modality treatments had minor effects. For MCa-35, radiation or the combination led to equivalent growth inhibition. In all tumors, hypoxia increased markedly after either radiation or DC101 alone. Although combination therapy produced no immediate pathophysiological changes, hypoxia ultimately increased after cessation of therapy. Preferential increases in endothelial apoptosis following combination treatment suggest that in addition to blocking tumor angiogenesis, DC101 enhances radiotherapy by specifically sensitizing endothelial cells, leading to degeneration of newly formed blood vessels.


Subject(s)
Antibodies, Monoclonal/pharmacology , Mammary Neoplasms, Experimental/radiotherapy , Mammary Neoplasms, Experimental/therapy , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Cell Division/physiology , Cell Division/radiation effects , Cell Hypoxia/physiology , Cell Hypoxia/radiation effects , Combined Modality Therapy , Dose Fractionation, Radiation , Endothelium, Vascular/pathology , Endothelium, Vascular/radiation effects , Female , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/immunology , Mice , Mice, Inbred C3H , Vascular Endothelial Growth Factor Receptor-2/immunology
14.
Radiother Oncol ; 72(2): 221-30, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15297140

ABSTRACT

BACKGROUND AND PURPOSE: The primary objectives of this study were to address two major questions. (1) Does VEGF receptor-2 antibody (DC101) produce detrimental effects on tumor vascular function and oxygenation that could compromise adjuvant therapies? (2) Is pathophysiological response to such antiangiogenic strategies different in transplanted versus primary spontaneous tumors? MATERIALS AND METHODS: The effects of early and late initiation DC101 treatment were evaluated using spontaneous murine mammary carcinomas and two markedly different transplanted mammary tumors, MCa-35 and MCa-4. Mice were administered DC101 or saline, tumors were frozen, and immunohistochemical staining was quantified using image analysis of multiply-stained frozen sections. Total blood vessels were identified using antibodies to CD31 or panendothelial antigen, perfused vessels via i.v. injection of fluorescent DiOC7, and tumor hypoxia by hypoxia marker (EF5) uptake. RESULTS: Tumor growth was significantly inhibited following DC101 administration in all tumor models. In general, early initiation DC101 treatment reduced perfused vessel counts and increased tumor hypoxia, while late initiation treatment had no significant impact on either. Results indicate that DC101 slows tumor growth through a decrease in vascular function, leading to increased tumor cell apoptosis and necrosis at sites distant from perfused blood vessels, and suggest that DC101 accelerates the rate at which tumor cells outgrow their functional vascular supply. CONCLUSIONS: Although highly variable among individual spontaneous tumors, the overall effects of DC101 on tumor hypoxia were quite similar between spontaneous and transplanted tumors. Since reductions in tumor oxygenation due to antiangiogenic treatment were transient, initial pathophysiological deficiencies that could compromise conventional therapies over the short-term may be of less relevance when administered over more extended treatment schedules.


Subject(s)
Breast Neoplasms/immunology , Cell Hypoxia/immunology , Endothelium, Vascular/immunology , Mammary Neoplasms, Animal/blood supply , Mammary Neoplasms, Animal/immunology , Vascular Endothelial Growth Factor Receptor-2/immunology , Animals , Antibodies/administration & dosage , Cell Line, Tumor , Female , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/immunology , Mice , Mice, Inbred C3H , Neoplasm Transplantation
15.
Adv Exp Med Biol ; 530: 165-76, 2003.
Article in English | MEDLINE | ID: mdl-14562714

ABSTRACT

Since quantitative measurements of tumor vascular function cannot be obtained in human tumors, appropriate animal tumor models must be utilized. The current studies were undertaken to compare transplantable, murine KHT tumors with primary and 1st generation transplants of spontaneous mammary carcinomas. To evaluate changes in tumor vascular structure and function, immunostaining of total and perfused vascular spacing, and cryospectrophotometric measurement of intravascular HbO2 saturations were utilized. KHT tumors demonstrated a distinct pattern of decreasing oxygenation with increasing distance from the tumor surface, while spontaneous tumors exhibited striking intertumor heterogeneities and a reduced dependence of oxygenation on distance from tumor surface. Anatomical/perfused vessel distributions and functional response were similar between the primary and transplanted tumor models, as was tissue histological appearance, but were quite different from KHT tumors. These results indicate that spontaneous tumor vascular configuration and function tend to be preserved in 1st generation trochar transplanted tumors.


Subject(s)
Mammary Neoplasms, Experimental/metabolism , Oxygen/metabolism , Animals , Immunohistochemistry , Mammary Neoplasms, Experimental/blood supply , Mice , Mice, Inbred C3H , Neoplasm Transplantation , Regional Blood Flow
16.
Adv Exp Med Biol ; 530: 593-601, 2003.
Article in English | MEDLINE | ID: mdl-14562756

ABSTRACT

Breast tumors expressing no detectable FGFs (MCF-7) were compared with tumors transfected with FGF4 or FGF1 (FGF4/MCF-7 or FGF1/MCF-7), and with MDA-MB-435, which produce endogenous FGF2. Tumor blood flow was measured by 133Xe diffusion, oxygen distribution was measured by Eppendorf pO2 histography, and vascular density was measured by CD31 staining. Tumors that overexpress angiogenic factors grew at a rate far exceeding that of MCF-7. The FGF producing tumors also had much higher metastatic rates to lung. Tumor blood flow was significantly higher in the two FGF-transfected xenografts compared with the parent MCF7. Median tumor pO2 was also higher, and tumor oxygenation was preserved even for large tumors. The vascular density as determined by CD31 staining, however, was not markedly increased in tumors overexpressing angiogenic factors. We found that angiogenic factors preserve and augment neovascular function, thus facilitating tumor growth and progression.


Subject(s)
Breast Neoplasms/pathology , Fibroblast Growth Factors/physiology , Neoplasm Metastasis , Oxygen/metabolism , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/metabolism , Cell Division , Cell Line, Tumor , Fibroblast Growth Factors/biosynthesis , Humans , Immunohistochemistry , Mice , Mice, Nude , Regional Blood Flow
17.
Adv Exp Med Biol ; 530: 603-9, 2003.
Article in English | MEDLINE | ID: mdl-14562757

ABSTRACT

Isotransplants of murine fibrosarcoma (KHT) cells were inoculated i.m. into the hind limbs of 6-8 week-old female C3H/HeJ mice. Intratumoral injection of FGF1 or VEGF proteins decreased hypoxic marker uptake in murine fibrosarcoma KHT. Reduction of tumor hypoxia did not correlate with mRNA expression of transcription factors in tumors. Likewise, there was no significant alteration in either apoptotic frequency or the mRNA levels of 10 apoptotic-related molecules in FGF1- or VEGF-treated tumors. mRNA expression for MCP-1, IL-1 beta, IL-18, and IL-1Ra, however, were decreased in the tumors following FGF1 or VEGF treatment. Among the normal tissues tested (brain, kidney, liver, spleen, and lung), basal mRNA levels for cytokines and chemokines varied. Intratumoral injection of FGF1 or VEGF (6 daily intra-tumor injections of 6 micrograms/mouse) did not alter most cytokine or chemokine mRNA expression in spleen and lung. In summary, alteration of tumor oxygenation by local administration of angiogenic growth factors may be mediated by cytokine/chemokine production in the tumor.


Subject(s)
Fibroblast Growth Factor 1/physiology , Fibrosarcoma/pathology , Neovascularization, Pathologic , Vascular Endothelial Growth Factor A/physiology , Animals , Cell Line, Tumor , Female , Mice , Mice, Inbred C3H , Neoplasm Transplantation
18.
Int J Radiat Oncol Biol Phys ; 57(4): 1038-46, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-14575835

ABSTRACT

PURPOSE: Recent results in the literature have demonstrated that the antiangiogenic agent endostatin can enhance antitumor effects when administered before or during radiotherapy. To better understand the underlying pathophysiologic basis for this radiosensitization, the current study investigated whether short-term endostatin administration is linked to alterations in tumor vascular perfusion and oxygen delivery. METHODS AND MATERIALS: Three daily doses of recombinant endostatin (20 mg/kg) were administered to two murine mammary carcinomas, the highly vascularized MCa-35 and the less vascularized MCa-4. Image analysis techniques were used to quantify (1) total and perfused vascular spacing, and (2) changes in tumor hypoxia as a function of distance from the nearest blood vessel. RESULTS: In MCa-35 tumors, endostatin had no effect on vessel spacing, tumor hypoxia, or tumor growth. In MCa-4 tumors, total and perfused vessel spacings were also unchanged, but tumor growth was inhibited, and tumor hypoxia significantly decreased. These tumors demonstrated an increased vascular functionality suggestive of an increase in the number of intermittently perfused vessels, without corresponding alterations in tumor oxygen consumption rate. CONCLUSIONS: Poorly vascularized, hypoxic mammary carcinomas were much more responsive to short-term endostatin treatment than well-vascularized, more homogeneously oxygenated tumors. Oxygen levels in the responsive tumors were transiently improved after treatment, which could have substantial implications with respect to the therapeutic effectiveness of combining antiangiogenic agents with conventional therapies.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Cell Hypoxia/drug effects , Endostatins/pharmacology , Mammary Neoplasms, Experimental/blood supply , Neovascularization, Pathologic/drug therapy , Animals , Apoptosis , Carbocyanines , Cell Division , Drug Evaluation, Preclinical , Female , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C3H
19.
Cancer Res ; 63(15): 4684-91, 2003 Aug 01.
Article in English | MEDLINE | ID: mdl-12907650

ABSTRACT

Alteration of the phenotype of breast cancers from estrogen-dependent to estrogen-independent growth often leads to the failure of antiestrogenic tumor therapies. We report that overexpression of vascular endothelial growth factor (VEGF) by estrogen-dependent MCF-7 breast cancer cells could abolish estrogen-dependent tumor growth in ovariectomized mice. In the absence of estrogen, MCF-7 VEGF-expressing tumors with increased vessel density showed growth kinetics similar to, or even greater than, that of parental MCF-7 tumors with estrogen supplementation. Overexpression of VEGF by MCF-7 cells or treatment on parental MCF-7 cells with recombinant VEGF also stimulated cell proliferation in culture. Our data suggest that VEGF stimulation of MCF-7 tumor angiogenesis and growth is mediated by both autocrine and paracrine mechanisms.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Endothelial Growth Factors/biosynthesis , Estradiol/pharmacology , Intercellular Signaling Peptides and Proteins/biosynthesis , Lymphokines/biosynthesis , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/genetics , Cell Division/drug effects , Cell Division/physiology , Endothelial Growth Factors/genetics , Female , Humans , Intercellular Signaling Peptides and Proteins/genetics , Lac Operon , Lymphokines/genetics , Mice , Mice, Nude , Neoplasms, Hormone-Dependent/blood supply , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Protein Isoforms , Receptors, Vascular Endothelial Growth Factor/biosynthesis , Transfection , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...