Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Front Oncol ; 9: 247, 2019.
Article in English | MEDLINE | ID: mdl-31024849

ABSTRACT

Introduction: Metformin, the most widely used treatment for diabetes, is lethal to cancer cells and increases in toxicity when used in combination with radiation. In addition to various molecular and metabolic mechanisms that have been previously proposed, the studies presented provide evidence of an additional, novel mechanism of sensitization following high dose radiotherapy; the magnitude of sensitization depends on the microenvironmental levels of glucose and oxygen which are in turn affected by high dose radiation. Methods: Cancer cells (A549 and MCF7) were studied in vitro under various controlled conditions. Endpoints included clonogenic cell survival and ROS expression measured by DHE and DCFDA. CD1 nu/nu athymic mice implanted with A549 cells received metformin alone (200 mg/kg, i.p.), radiation alone (15 Gy) or a combination of metformin and radiation; the effect of treatment sequence on efficacy was assessed by tumor growth delay and histology. In a separate set of experiments, tumor blood flow was measured using a tracer clearance technique using SPECT after the administration of metformin alone, radiation alone and the combined treatment. Results: In vivo, metformin provided equally effective tumor growth delay when given 24 h after radiation as when given 1 h or 4 h before radiation, an observation not previously reported and, in fact, unexpected based on published scientific literature. When drug followed radiation, the tumors were histologically characterized by massive cellular necrosis. In vitro, cancer cells when glucose depleted and/or hypoxic were preferentially killed by metformin, in a drug dose dependent manner. A549 cells exposed to 5.0 mM of metformin was reduced seven fold in survival when in a glucose deprived as compared to a low-glucose medium (0 vs. 1.0 g/L). Finally, using a SPECT detector to follow the washout of a radioactive tracer, it was shown that a high single dose of radiosurgery (15 Gy) could dramatically inhibit blood flow and presumably diminish glucose and oxygen. Discussion: Insight into the best timing of drug and radiation administration is gained through an understanding of the mechanisms of interaction. A new mechanism of metformin sensitization by high dose radiation is proposed based on the blood flow, glucose and oxygen.

2.
Radiat Oncol J ; 36(3): 172-181, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30309208

ABSTRACT

Successful anticancer strategies require a differential response between tumor and normal tissue (i.e., a therapeutic ratio). In fact, improving the effectiveness of a cancer therapeutic is of no clinical value in the absence of a significant increase in the differential response between tumor and normal tissue. Although radiation dose escalation with the use of intensity modulated radiation therapy has permitted the maximum tolerable dose for most locally advanced cancers, improvements in tumor control without damaging normal adjacent tissues are needed. As a means of increasing the therapeutic ratio, several new approaches are under development. Drugs targeting signal transduction pathways in cancer progression and more recently, immunotherapeutics targeting specific immune cell subsets have entered the clinic with promising early results. Radiobiological research is underway to address pressing questions as to the dose per fraction, irradiated tumor volume and time sequence of the drug administration. To exploit these exciting novel strategies, a better understanding is needed of the cellular and molecular pathways responsible for both cancer and normal tissue and organ response, including the role of radiation-induced accelerated senescence. This review will highlight the current understanding of promising biologically targeted therapies to enhance the radiation therapeutic ratio.

3.
Front Behav Neurosci ; 12: 87, 2018.
Article in English | MEDLINE | ID: mdl-29867388

ABSTRACT

Mice socially isolated during adolescence exhibit behaviors of anxiety, depression and impaired social interaction. Although these behaviors are well documented, very little is known about the associated neurobiological changes that accompany these behaviors. It has been hypothesized that social isolation during adolescence alters the development of the prefrontal cortex, based on similar behavioral abnormalities observed in isolated mice and those with disruption of this structure. To establish relationships between behavior and underlying neurobiological changes in the prefrontal cortex, Thy-1-GFP mice were isolated from weaning until adulthood and compared to group-housed littermates regarding behavior, electrophysiological activity and dendritic morphology. Results indicate an immaturity of dendritic spines in single housed animals, with dendritic spines appearing smaller and thinner. Single housed mice additionally show impaired plasticity through measures of long-term potentiation. Together these findings suggest an altered development and impairment of the prefrontal cortex of these animals underlying their behavioral characteristics.

5.
Radiat Oncol J ; 32(3): 103-15, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25324981

ABSTRACT

To summarize current knowledge regarding mechanisms of radiation-induced normal tissue injury and medical countermeasures available to reduce its severity. Advances in radiation delivery using megavoltage and intensity-modulated radiation therapy have permitted delivery of higher doses of radiation to well-defined tumor target tissues. Injury to critical normal tissues and organs, however, poses substantial risks in the curative treatment of cancers, especially when radiation is administered in combination with chemotherapy. The principal pathogenesis is initiated by depletion of tissue stem cells and progenitor cells and damage to vascular endothelial microvessels. Emerging concepts of radiation-induced normal tissue toxicity suggest that the recovery and repopulation of stromal stem cells remain chronically impaired by long-lived free radicals, reactive oxygen species, and pro-inflammatory cytokines/chemokines resulting in progressive damage after radiation exposure. Better understanding the mechanisms mediating interactions among excessive generation of reactive oxygen species, production of pro-inflammatory cytokines and activated macrophages, and role of bone marrow-derived progenitor and stem cells may provide novel insight on the pathogenesis of radiation-induced injury of tissues. Further understanding the molecular signaling pathways of cytokines and chemokines would reveal novel targets for protecting or mitigating radiation injury of tissues and organs.

6.
Radiat Res ; 182(3): 316-21, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25098729

ABSTRACT

Radiation injury to skin poses substantial morbidity risks in the curative treatment of cancers and is also of concern in the context of radiological attack or nuclear accident scenarios. Late effects can be severe and are frequently characterized by subcutaneous fibrosis and morbidity. These experiments presented here assess the potential of MW01-2-151SRM (MW-151), a novel small-molecule inhibitor of microglial activation and associated proinflammatory cytokine/chemokine production, as a mitigator of radiation-induced skin injury. Groups of C57BL/6 mice received focal irradiation of the right hind leg at a dose of 30 Gy. Therapy was initiated either on day 3, day 7 or day 14 postirradiation and maintained subsequently for 21 days by intraperitoneal injections administered three times per week. The primary end point was skin injury, which was assessed three times a week for at least 60 days postirradiation and scored using a semi-quantitative scale. Secondary end points measured at selected times included histology (primarily H&E) and immunofluorescence labeling of various macrophage (F4-80) and inflammatory (TGF-ß, TNF-α, MMP9) markers. Relative to untreated controls, mitigation of radiation-induced skin injury in mice receiving MW-151 was highly dependent on the timing of therapy initiation. Initiation on day 3 postirradiation had no discernable effect, whereas mitigating effects were maximal following initiation on day 7 and present to a lesser degree following initiation on day 14. The response to MW-151 therapy in individual animals was essentially all-or-none and the relative benefits associated with the timing of therapy initiation primarily reflected differences in the number of responders. These data support the hypothesis that proinflammatory cytokines/chemokines play complex roles in orchestrating the response to radiation-induced skin injury and suggest that there is a critical period during which they initiate the pathogenesis resulting in late effects.


Subject(s)
Cytokines/physiology , Pyridazines/pharmacology , Pyrimidines/pharmacology , Radiation Injuries/prevention & control , Skin/radiation effects , Animals , Cytokines/antagonists & inhibitors , Macrophages/drug effects , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Radiation Injuries/immunology , Radiation Injuries/pathology , Skin/pathology , Time Factors
7.
Radiat Res ; 179(5): 549-56, 2013 May.
Article in English | MEDLINE | ID: mdl-23560629

ABSTRACT

Cognitive impairment precipitated by irradiation of normal brain tissue is commonly associated with radiation therapy for treatment of brain cancer, and typically manifests more than 6 months after radiation exposure. The risks of cognitive impairment are of particular concern for an increasing number of long-term cancer survivors. There is presently no effective means of preventing or mitigating this debilitating condition. Neuroinflammation mediated by activated microglial cytokines has been implicated in the pathogenesis of radiation-induced cognitive impairment in animal models, including the disruption of neurogenesis and activity-induced gene expression in the hippocampus. These pathologies evolve rapidly and are associated with relatively subtle cognitive impairment at 2 months postirradiation. However, recent reports suggest that more profound cognitive impairment develops at later post-irradiation time points, perhaps reflecting a gradual loss of responsiveness within the hippocampus by the disruption of neurogenesis. We hypothesized that inhibiting neuroinflammation using MW01-2-151SRM (MW-151), a selective inhibitor of proinflammatory cytokine production, might mitigate these deleterious radiation effects by preserving/restoring hippocampal neurogenesis. MW-151 therapy was initiated 24 h after 10 Gy whole-brain irradiation (WBI) administered as a single fraction and maintained for 28 days thereafter. Proinflammatory activated microglia in the dentate gyrus were assayed at 2 and 9 months post-WBI. Cell proliferation and neurogenesis in the dentate gyrus were assayed at 2 months post-WBI, whereas novel object recognition and long-term potentiation were assayed at 6 and 9 months post-WBI, respectively. MW-151 mitigated radiation-induced neuroinflammation at both early and late time points post-WBI, selectively mitigated the deleterious effects of irradiation on hippocampal neurogenesis, and potently mitigated radiation-induced deficits of novel object recognition consolidation and of long-term potentiation induction and maintenance. Our results suggest that transient administration of MW-151 is sufficient to partially preserve/restore neurogenesis within the subgranular zone and to maintain the functional integrity of the dentate gyrus long after MW-151 therapy withdrawal.


Subject(s)
Cognition Disorders/drug therapy , Cognition Disorders/pathology , Microglia/pathology , Pyridazines/pharmacology , Pyrimidines/pharmacology , Radiation Injuries, Experimental/drug therapy , Radiation Injuries, Experimental/pathology , Animals , Cognition Disorders/metabolism , Cognition Disorders/physiopathology , Cytokines/biosynthesis , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiopathology , Hippocampus/radiation effects , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Inflammation/physiopathology , Long-Term Potentiation/drug effects , Long-Term Potentiation/radiation effects , Male , Microglia/drug effects , Microglia/metabolism , Microglia/radiation effects , Neurogenesis/drug effects , Neurogenesis/radiation effects , Pyridazines/therapeutic use , Pyrimidines/therapeutic use , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/physiopathology , Rats , Rats, Inbred F344 , Recognition, Psychology/drug effects , Recognition, Psychology/radiation effects , Whole-Body Irradiation/adverse effects
8.
Int J Radiat Biol ; 89(5): 311-8, 2013 May.
Article in English | MEDLINE | ID: mdl-23305180

ABSTRACT

PURPOSE: To summarize current knowledge regarding mechanisms of radiation-induced skin injury and medical countermeasures available to reduce its severity. Advances in radiation delivery using megavoltage and intensity modulated radiation therapy have permitted delivery of higher doses of radiation to well-defined tumor target tissues. Although skin is not a radiation dose-limiting tissue, injury to skin poses substantial morbidity risks in the curative treatment of cancers, especially when radiation is administered in combination with chemotherapy. In the continuum of radiation-induced skin injury, late effects are most severe being characterized by sub-cutaneous fibrosis and morbidity. The principal pathogenesis is initiated by depletion of acutely responding epithelial tissues and damage to vascular endothelial microvessels. Emerging concepts of radiation- induced skin injury suggest that the recovery of stromal stem cells and tissue repair remain chronically impaired by long-lived free radicals, reactive oxygen species, and pro-inflammatory cytokines/chemokines resulting in progressive damage after radiation exposure. CONCLUSIONS: As pathways underlying the cellular and molecular mechanisms of radiation-induced skin injury are becoming better understood, novel approaches are being developed for mitigating or treating the associated pathogenesis.


Subject(s)
Clinical Trials as Topic/methods , Radiation Injuries , Skin/injuries , Skin/radiation effects , Animals , Bone Marrow Cells/radiation effects , Chemokines/biosynthesis , Humans , Radiation Injuries/immunology , Radiation Injuries/metabolism , Radiation Injuries/therapy , Reactive Oxygen Species/metabolism , Skin/immunology , Skin/metabolism
9.
Radiat Res ; 178(3): 202-6, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22799631

ABSTRACT

Even with modern 3D conformal treatments skin radiation injury can be an inadvertent complication associated with clinical radiotherapy particularly at tissue folds. It is also of concern in the context of a radiological terrorism incident or accident, since skin irradiation lowers the lethal dose of whole body radiation. We hypothesize that radiation-induced skin injury originates from a loss of stem and progenitor cells, accompanied by excessive ROS production and proinflammatory cytokines. Plerixafor, a CXCR-4 antagonist, is one of the most efficient bone marrow stem cell mobilizers and these studies were designed to experimentally assess the potential of Plerixafor to reduce skin radiation injury. The right hind legs of groups of C57BL/6 mice were exposed to radiation alone or in combination with Plerixafor. Plerixafor was administered intraperitoneally at a dose of 5 mg/kg given in two doses separated by two days and started either on day 0, 4, 7, 15 or 24 after irradiation. The primary end point was skin injury, which was assessed three times a week for at least 2 months using a semi-quantitative scale. Secondary end points measured at selected time points included histology (primarily H&E) and cytokine levels (TGF-ß and TNF-α). The acute and late skin injury in mice receiving Plerixafor was highly dependent on the timing of administration of the drug. The maximum benefit was observed when the drug was started 1 week after radiation exposure, and earlier or later administration of the drug decreased its efficacy. Secondary damage end points (cytokine levels and histologically assessed tissue thickness) provided confirmatory observations. In an attempt to gain insight into the effect of timing of administration of the agent on the mitigation effect, the ligand to CXCR4, stromal derived factor, SDF-1, was measured as a function of time after radiation exposure. Expression of SDF-1 monitored in skin as a function of time after a 30 Gy radiation exposure suggested a strong correlation between timing of administration of Plerixafor and expression of SDF-1 in irradiated skin: optimum drug administration timing coincided with maximal SDF-1 expression in the skin of irradiated mice. This report presents the first observation that CXCR4 antagonist improves both acute and late skin response to radiation exposure.


Subject(s)
Heterocyclic Compounds/pharmacology , Radiation Injuries, Experimental/drug therapy , Radiation-Protective Agents/pharmacology , Receptors, CXCR4/antagonists & inhibitors , Skin/drug effects , Skin/injuries , Animals , Benzylamines , Cyclams , Dose-Response Relationship, Radiation , Heterocyclic Compounds/administration & dosage , Heterocyclic Compounds/therapeutic use , Mice , Mice, Inbred C57BL , Radiation Injuries, Experimental/pathology , Radiation-Protective Agents/administration & dosage , Radiation-Protective Agents/therapeutic use , Skin/pathology , Skin/radiation effects , Time Factors
10.
Fluids Barriers CNS ; 9(1): 13, 2012 Jul 11.
Article in English | MEDLINE | ID: mdl-22784705

ABSTRACT

BACKGROUND: Hydrocephalus is a central nervous system (CNS) disorder characterized by the abnormal accumulation of cerebrospinal fluid (CSF) in cerebral ventricles, resulting in their dilatation and associated brain tissue injury. The pathogenesis of hydrocephalus remains unclear; however, recent reports suggest the possible involvement of abnormal osmotic gradients. Here we explore the kinetics associated with manipulating CSF osmolarity on ventricle volume (VV) in the normal rat brain. METHODS: CSF was made hyper-osmotic by introducing 10KD dextran into the lateral ventricle, either by acute injection at different concentrations or by chronic infusion at a single concentration. The induction and withdrawal kinetics of dextran infusion on VV were explored in both contexts. RESULTS: Acute intraventricular injection of dextran caused a rapid increase in VV which completely reversed within 24 hours. These kinetics are seemingly independent of CSF osmolarity across a range spanning an order of magnitude; however, the magnitude of the transient increase in VV was proportional to CSF osmolarity. By contrast, continuous intraventricular infusion of dextran at a relatively low concentration caused a more gradual increase in VV which was very slow to reverse when infusion was suspended after five days. CONCLUSION: We conclude that hyperosmolar CSF is sufficient to produce a proportional degree of hydrocephalus in the normal rat brain, and that this phenomenon exhibits hysteresis if CSF hyperosmolarity is persistent. Thus pathologically-induced increases in CSF osmolarity may be similarly associated with certain forms of clinical hydrocephalus. An improved understanding of this phenomenon and its kinetics may facilitate the development of novel therapies for the treatment of clinical hydrocephalus.

11.
J Neurooncol ; 101(3): 449-56, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20617366

ABSTRACT

Whole brain irradiation (WBI) is commonly administered therapeutically and is routinely associated with late delayed radiation injuries, manifesting as severe and irreversible cognitive impairment. Neural progenitors within the subgranular zone (SGZ) of the dentate gyrus are among the most radiosensitive cell types in the adult brain and are known to participate in hippocampal plasticity and normal cognitive function. These progenitors and the specialized SGZ microenvironment required for neuronal differentiation are the source of neurogenic potential in the adult dentate gyrus, and provide a continuous supply of immature neurons which may then migrate into the adjacent granule cell layer to become mature granule cell neurons. The extreme radiosensitivity of these progenitors and the SGZ microenvironment implicate them as potentially significant contributors to radiation-induced cognitive impairment. Previous reports suggest that statin drugs may be neuroprotective and may promote neurogenesis within the SGZ following both traumatic and ischemic brain injury. Here we investigate whether atorvastatin might similarly protect progenitors and/or preserve neurogenic potential within the SGZ when administered following radiation injury. We also investigate whether such mitigating effects might be enhanced by administering atorvastatin in combination with the angiotensin converting enzyme (ACE) inhibitor, ramipril, which has previously been shown to produce subtle mitigating effects in this context. Atorvastatin was administered to adult male Fisher 344 rats beginning 24 h post-WBI at doses of 10 and 15 Gy, and maintained daily until sacrifice at 12 weeks post-WBI. Combined atorvastatin and ramipril (atorvastatin + ramipril) were administered according to the same protocol following WBI doses of 10 Gy. Progenitor proliferation, neuronal differentiation, and microglial activation were assayed immunohistochemically. Our results indicate that chronic administration of atorvastatin is relatively ineffective as a mitigator of radiation injury in this context, whereas atorvastatin + ramipril appear to interact synergistically to potently and selectively mitigate radiation-induced disruption of neurogenic signaling within SGZ microenvironment.


Subject(s)
Cranial Irradiation/adverse effects , Dentate Gyrus/drug effects , Heptanoic Acids/therapeutic use , Neurogenesis/drug effects , Pyrroles/therapeutic use , Radiation Injuries, Experimental/prevention & control , Ramipril/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Anticholesteremic Agents/therapeutic use , Atorvastatin , Cesium Radioisotopes , Dentate Gyrus/radiation effects , Immunoenzyme Techniques , Male , Neurogenesis/radiation effects , Radiation Injuries, Experimental/etiology , Rats , Rats, Inbred F344
12.
Radiat Res ; 173(4): 462-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20334518

ABSTRACT

Antioxidants mitigate radiation-induced lethality when started soon after radiation exposure, a delivery time that may not be practical due to difficulties in distribution and because the oral administration of such agents may require a delay beyond the prodromal stage of the radiation syndrome. We report the unexpected finding that antioxidant supplementation starting 24 h after total-body irradiation resulted in better survival than antioxidant supplementation started soon after the irradiation. The antioxidant dietary supplement was l-selenomethionine, sodium ascorbate, N-acetyl cysteine, alpha-lipoic acid, alpha-tocopherol succinate, and co-enzyme Q10. Total-body irradiation with 8 Gy in the absence of antioxidant supplementation was lethal by day 16. When antioxidant supplementation was started soon after irradiation, four of 14 mice survived. In contrast, 14 of 18 mice receiving antioxidant supplementation starting 24 h after irradiation were alive and well 30 days later. The numbers of spleen colonies and blood cells were higher in mice receiving antioxidant supplementation starting 24 h after irradiation than in mice receiving radiation alone. A diet supplemented with antioxidants administered starting 24 h after total-body irradiation improved bone marrow cell survival and mitigated lethality, with a radiation protection factor of approximately 1.18.


Subject(s)
Antioxidants/therapeutic use , Radiation Injuries/mortality , Radiation Injuries/prevention & control , Radiation Protection/methods , Survival Rate , Whole-Body Irradiation/statistics & numerical data , Animals , Dietary Supplements , Dose-Response Relationship, Radiation , Mice , Mice, Inbred C57BL , Prevalence , Radiation Injuries/veterinary , Survival Analysis
13.
Radiat Oncol ; 5: 6, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20122169

ABSTRACT

BACKGROUND: Sublethal doses of whole brain irradiation (WBI) are commonly administered therapeutically and frequently result in late delayed radiation injuries, manifesting as severe and irreversible cognitive impairment. Neural progenitors within the subgranular zone (SGZ) of the dentate gyrus are among the most radiosensitive cell types in the adult brain and are known to participate in hippocampal plasticity and normal cognitive function. These progenitors and the specialized SZG microenvironment required for neuronal differentiation are the source of neurogenic potential in the adult dentate gyrus, and provide a continuous supply of immature neurons which may then migrate into the adjacent granule cell layer to become mature granule cell neurons. The extreme radiosensitivity of these progenitors and the SGZ microenvironment suggests the hippocampus as a prime target for radiation-induced cognitive impairment. The brain renin-angiotensin system (RAS) has previously been implicated as a potent modulator of neurogenesis within the SGZ and selective RAS inhibitors have been implicated as mitigators of radiation brain injury. Here we investigate the angiotensin converting enzyme (ACE) inhibitor, ramipril, as a mitigator of radiation injury in this context. METHODS: Adult male Fisher 344 rats received WBI at doses of 10 Gy and 15 Gy. Ramipril was administered beginning 24 hours post-WBI and maintained continuously for 12 weeks. RESULTS: Ramipril produced small but significant reductions in the deleterious effects of radiation on progenitor proliferation and neuronal differentiation in the rat dentate gyrus following 10 Gy-WBI, but was not effective following 15 Gy-WBI. Ramipril also reduced the basal rate of neurogenesis within the SGZ in unirradiated control rats. CONCLUSIONS: Our results indicate that chronic ACE inhibition with ramipril, initiated 24 hours post-irradiation, may reduce apoptosis among SGZ progenitors and/or inflammatory disruption of neurogenic signaling within SGZ microenvironment, and suggest that angiotensin II may participate in maintaining the basal rate of granule cell neurogenesis.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Cranial Irradiation/adverse effects , Dentate Gyrus/radiation effects , Neurogenesis/radiation effects , Radiation Injuries, Experimental/prevention & control , Ramipril/therapeutic use , Animals , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Immunohistochemistry , Male , Neurons/radiation effects , Radiation Injuries, Experimental/etiology , Rats , Rats, Inbred F344 , Stem Cells/radiation effects
14.
J Neurooncol ; 87(3): 279-86, 2008 May.
Article in English | MEDLINE | ID: mdl-18209952

ABSTRACT

Radiation therapy is widely used in the treatment of primary malignant brain tumors and metastatic tumors of the brain with either curative or palliative intent. The limitation of cancer radiation therapy does not derive from the inability to ablate tumor, but rather to do so without excessively damaging the patient. Among the varieties of radiation-induced brain toxicities, it is the late delayed effects that lead to severe and irreversible neurological consequences. Following radiation exposure, late delayed effects within the CNS have been attributable to both parenchymal and vascular damage involving oligodendrocytes, neural progenitors, and endothelial cells. These reflect a dynamic process involving radiation-induced death of target cells and subsequent secondary reactive neuroinflammatory processes that are believed to lead to selective cell loss, tissue damage, and functional deficits. The progressive, late delayed damage to the brain after high-dose radiation is thought to be caused by radiation-induced long-lived free radicals, reactive oxygen species, and pro-inflammatory cytokines. Experimental studies suggest that radiation-induced brain injury can be successfully mitigated and treated with several well established drugs in wide clinical use which exert their effects by blocking pro-inflammatory cytokines and reactive oxygen species. This review highlights preclinical and early clinical data that are translatable for future clinical trials.


Subject(s)
Brain Neoplasms/radiotherapy , Brain/radiation effects , Clinical Trials as Topic , Radiation Injuries/prevention & control , Radiotherapy/adverse effects , Animals , Humans , Radiation Injuries/etiology
15.
J Neurooncol ; 82(2): 119-24, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17004100

ABSTRACT

PURPOSE: Radiation-induced optic nerve damage was reduced by ramipril, a prodrug angiotensin-converting enzyme inhibitor (ACEI). This study was to determine the optimum dose and administration time of ramipril for mitigating radiation-induced optic neuropathy. MATERIALS AND METHOD: Adult Fischer 344 male rats were treated with a single dose radiation 30 Gy by using radiosurgical technique. After irradiation, the animals were randomly assigned into groups of different ramipril doses and administration time; control (no treatment), radiation alone, radiation+ramipril in different doses and starting times of drug. Ramipril was given 0.5-1.5 mg/kg/day and AT1R blocker Losartan 20 mg/kg/day in drinking water for 180 days. Functional endpoint with visual evoked potential (VEP) and anatomical endpoint with gross and histological analysis with immunohistochemical (IHC) stain were used. RESULTS: Normal VEP measurements in un-irradiated rats were 46.2+/-7.9 ms. There was no change of VEP value until 4 months, but was lengthened to 188.1+/-58.7 ms at 6 months after radiation. By ramipril treatment with the dose of 1.5 mg starting at 2 weeks after radiation, VEP was significantly shortened to 105.7+/-88.5 ms at 6 months. Gross and microscopic structure of the irradiated optic nerve was well preserved in the ramipril-treated group. CONCLUSION: Ramipril can mitigate the radiation-induced optic nerve damage and preserve the functional integrity of the nerve. The results support early treatment with a high dose of ramipril after radiation.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Optic Nerve Diseases/drug therapy , Optic Nerve/radiation effects , Radiation Injuries, Experimental/drug therapy , Radiation-Protective Agents/administration & dosage , Ramipril/administration & dosage , Animals , Dose-Response Relationship, Radiation , Evoked Potentials, Visual/radiation effects , Injections, Intraperitoneal , Male , Optic Nerve Diseases/etiology , Radiation Injuries, Experimental/etiology , Rats , Rats, Inbred F344 , Survival Rate , Time Factors
16.
Brain Res ; 1094(1): 207-16, 2006 Jun 13.
Article in English | MEDLINE | ID: mdl-16762327

ABSTRACT

Several clinical and experimental reports suggest that low-dose irradiation of an established epileptic focus can reduce the occurrence of spontaneous seizures. Conversely, some recent reports suggest that under some conditions low-dose irradiation may have disinhibitory effects on seizure expression. Here, we have investigated mechanistic aspects of this phenomenon in the kindling model of epilepsy by applying focal irradiation at various points during kindling development. Rats were kindled to stage 5 by afterdischarge-threshold electrostimulation of the left amygdala. Treatment groups were irradiated using a collimated X-ray beam (18 MV) either prior to kindling, at kindling stage 3, or at kindling stage 5, by exposure of the left amygdala to a single-fraction central-axis dose of 25 Gy. Generalized seizure thresholds (GSTs) were subsequently assayed at weekly intervals for 10 weeks and at monthly intervals for an additional 3 months, along with the severity of the evoked seizures. Irradiation produced no significant effects on seizure threshold, but did produce persistent changes in seizure severity which varied as a function of the timing of irradiation. Relative to sham irradiated controls, the occurrence of stage 6 seizures was significantly increased by irradiation prior to kindling, but was unaffected by irradiation at kindling stage 3, and significantly reduced by irradiation at kindling stage 5. Quantitative immunohistochemical assays for neuron and astrocyte densities within the amygdala and hippocampus revealed only subtle changes in neuronal density within the dentate granule cell layer. These results are discussed in relation to mechanisms of seizure- and radiation-induced plasticity.


Subject(s)
Amygdala/radiation effects , Epilepsy/radiotherapy , Hippocampus/radiation effects , Kindling, Neurologic/radiation effects , Neural Pathways/radiation effects , Amygdala/pathology , Amygdala/physiopathology , Animals , Astrocytes/pathology , Astrocytes/radiation effects , Biomarkers/metabolism , Cell Count , DNA-Binding Proteins , Disease Models, Animal , Disease Progression , Electric Stimulation , Epilepsy/physiopathology , Epilepsy/prevention & control , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Kindling, Neurologic/physiology , Male , Nerve Tissue Proteins/metabolism , Neural Pathways/pathology , Neural Pathways/physiopathology , Neuronal Plasticity/physiology , Neuronal Plasticity/radiation effects , Neurons/pathology , Neurons/radiation effects , Nuclear Proteins/metabolism , Radiation Dosage , Radiation, Ionizing , Radiotherapy/methods , Rats , Rats, Wistar , Time Factors , Treatment Outcome
17.
Neurosci Lett ; 371(1): 45-50, 2004 Nov 16.
Article in English | MEDLINE | ID: mdl-15500964

ABSTRACT

Low-dose radiosurgery is presently in use as a treatment modality for focal epilepsy, but the mechanisms underlying the associated changes in seizure expression are poorly understood. We investigated whether total and parvalbumin expressing (PV+) neuronal densities within the hippocampus and amygdala are affected by analogous focal irradiation in amygdala-kindled rats. Adult rats were kindled by electrical stimulation through 10 stage 5 seizures. The kindled amygdala was then focally irradiated at 18 or 25 Gy, and generalized seizure thresholds were subsequently monitored for approximately 6 months. Histological and immunohistochemical assays of total and PV+ neuronal densities were performed bilaterally throughout the hippocampus and within the basolateral amygdala. PV+ neuronal densities were unaffected by kindling or irradiation in these regions. Kindling selectively reduced neuronal densities in the dentate granule cell layer, and medial CA3 pyramidal cell layer. Irradiation at 25 Gy, but not at 18 Gy, prevented or reversed this kindling-associated reduction in density.


Subject(s)
Dentate Gyrus/pathology , Dentate Gyrus/radiation effects , Epilepsy/radiotherapy , Kindling, Neurologic/radiation effects , Amygdala/physiopathology , Animals , Cell Count , Dentate Gyrus/physiopathology , Disease Models, Animal , Dose-Response Relationship, Radiation , Epilepsy/pathology , Epilepsy/physiopathology , Male , Parvalbumins/metabolism , Rats , Rats, Wistar
18.
Radiat Res ; 161(2): 137-42, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14731077

ABSTRACT

Inhibitors of angiotensin-converting enzyme (ACE) have been used to reduce radiation-induced normal tissue injury. The present study was carried out to determine whether ramipril, one of the inhibitors of ACE, would ameliorate radiation-induced brain damage, using a well-characterized optic neuropathy model in the rat, one of the most critical and radiosensitive structures in the brain. The brains of adult Fischer rats were irradiated stereotactically with 30 Gy using a single collimated beam. Six months after irradiation and 1.5 mg/kg day(-1) ramipril (started 2 weeks after irradiation), rats were assessed for optic nerve damage functionally, using visual evoked potential, and histologically. Results show that ramipril conferred significant modification of radiation injury, since rats receiving radiation alone showed a threefold lengthening in the mean peak latency in the visual evoked potential, whereas 75% of rats receiving radiation followed by ramipril had evoked potentials that resembled those of normal untreated control rats. The histology of irradiated and ramipril-treated optic nerves appeared nearly normal, while there was significant demyelination in both optic nerves of irradiated rats. The study represents the first demonstration of prophylaxis of radiation injury by a carboxyl-containing ACE inhibitor, providing a pharmacological strategy designed to reduce radiation-induced normal tissue damage.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Evoked Potentials, Visual/radiation effects , Optic Nerve/drug effects , Optic Nerve/radiation effects , Radiation Injuries, Experimental/drug therapy , Radiation Tolerance/drug effects , Radiation-Protective Agents/administration & dosage , Ramipril/administration & dosage , Administration, Oral , Animals , Dose-Response Relationship, Radiation , Male , Optic Nerve/pathology , Optic Nerve/physiopathology , Radiation Injuries, Experimental/prevention & control , Rats , Rats, Inbred F344 , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...