Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Antiño R. Allen is active.

Publication


Featured researches published by Antiño R. Allen.


PLOS ONE | 2012

Cranial Irradiation Alters Dendritic Spine Density and Morphology in the Hippocampus

Ayanabha Chakraborti; Antiño R. Allen; Barrett Allen; Susanna Rosi; John R. Fike

Therapeutic irradiation of the brain is a common treatment modality for brain tumors, but can lead to impairment of cognitive function. Dendritic spines are sites of excitatory synaptic transmission and changes in spine structure and number are thought to represent a morphological correlate of altered brain functions associated with hippocampal dependent learning and memory. To gain some insight into the temporal and sub region specific cellular changes in the hippocampus following brain irradiation, we investigated the effects of 10 Gy cranial irradiation on dendritic spines in young adult mice. One week or 1 month post irradiation, changes in spine density and morphology in dentate gyrus (DG) granule and CA1 pyramidal neurons were quantified using Golgi staining. Our results showed that in the DG, there were significant reductions in spine density at both 1 week (11.9%) and 1 month (26.9%) after irradiation. In contrast, in the basal dendrites of CA1 pyramidal neurons, irradiation resulted in a significant reduction (18.7%) in spine density only at 1 week post irradiation. Analysis of spine morphology showed that irradiation led to significant decreases in the proportion of mushroom spines at both time points in the DG as well as CA1 basal dendrites. The proportions of stubby spines were significantly increased in both the areas at 1 month post irradiation. Irradiation did not alter spine density in the CA1 apical dendrites, but there were significant changes in the proportion of thin and mushroom spines at both time points post irradiation. Although the mechanisms involved are not clear, these findings are the first to show that brain irradiation of young adult animals leads to alterations in dendritic spine density and morphology in the hippocampus in a time dependent and region specific manner.


Radiation Research | 2014

Exposure to Low-Dose 56Fe-Ion Radiation Induces Long-Term Epigenetic Alterations in Mouse Bone Marrow Hematopoietic Progenitor and Stem Cells

Isabelle R. Miousse; Lijian Shao; Jianhui Chang; Wei Feng; Yingying Wang; Antiño R. Allen; Jennifer Turner; Blair Stewart; Jacob Raber; Daohong Zhou; Igor Koturbash

There is an increasing need to better understand the long-term health effects of high-linear energy transfer (LET) radiation due to exposure during space missions, as well as its increasing use in clinical treatments. Previous studies have indicated that exposure to 56Fe heavy ions increases the incidence of acute myeloid leukemia (AML) in mice but the underlying molecular mechanisms remain elusive. Epigenetic alterations play a role in radiation-induced genomic instability and the initiation and progression of AML. In this study, we assessed the effects of low-dose 56Fe-ion irradiation on epigenetic alterations in bone marrow mononuclear cells (BM-MNCs) and hematopoietic progenitor and stem cells (HPSCs). Exposure to 56Fe ions (600 MeV, 0.1, 0.2 and 0.4 Gy) resulted in significant epigenetic alterations involving methylation of DNA, the DNA methylation machinery and expression of repetitive elements. Four weeks after irradiation, these changes were primarily confined to HPSCs and were exhibited as dose-dependent hypermethylation of LINE1 and SINE B1 repetitive elements [4.2-fold increase in LINE1 (P < 0.001) and 7.6-fold increase in SINE B1 (P < 0.01) after exposure to 0.4 Gy; n = 5]. Epigenetic alterations were persistent and detectable for at least 22 weeks after exposure, when significant loss of global DNA hypomethylation (1.9-fold, P < 0.05), decreased expression of Dnmt1 (1.9-fold, P < 0.01), and increased expression of LINE1 and SINE B1 repetitive elements (2.8-fold, P < 0.001 for LINE1 and 1.9-fold, P < 0.05 for SINE B1; n = 5) were observed after exposure to 0.4 Gy. In contrast, exposure to 56Fe ions did not result in accumulation of increased production of reactive oxygen species (ROS) and DNA damage, exhibited as DNA strand breaks. Furthermore, no significant alterations in cellular senescence and apoptosis were detected in HPSCs after exposure to 56Fe-ion radiation. These findings suggest that epigenetic reprogramming is possibly involved in the development of radiation-induced genomic instability and thus, may have a causative role in the development of AML.


Radiation Research | 2014

28Silicon Radiation-Induced Enhancement of Synaptic Plasticity in the Hippocampus of Naïve and Cognitively Tested Mice

Jacob Raber; Emil Rudobeck; Mary Campbell-Beachler; Antiño R. Allen; Barrett Allen; Susanna Rosi; Gregory A. Nelson; Jennifer Turner; John R. Fike; Roman Vlkolinsky

The space radiation environment consists of multiple species of high-energy charge particles (HZE), including 56Fe and 28Si nuclei, that may impact neuronal cells, but their damaging effects on the central nervous system (CNS) have been poorly defined. Hippocampus-dependent memory functions have been shown to be highly sensitive to 56Fe HZE particles, which poses a significant risk to the cognitive performance of astronauts during space missions. While low doses of 56Fe radiation do not induce cell death of mature neurons, they affect synaptic plasticity in the CA1 region, the principal neuronal output of the hippocampal formation involved in memory formation. The effects of 28Si on the CNS have not been defined. Compared to behaviorally naïve mice, cognitive testing might affect synaptic plasticity and the effects of 28Si radiation on synaptic plasticity might be modulated by prior cognitive testing. Therefore, in the current study, we quantified the effects of whole-body 28Si radiation (600 MeV/n, 0.25 and 1 Gy) on hippocampus-dependent contextual freezing and synaptic plasticity in the CA1 region of animals not exposed (behaviorally naïve mice) and animals exposed to the contextual freezing test (cognitively tested mice). In behaviorally naïve mice exposed to 0.25 and 1 Gy of 28Si radiation, the magnitude of long-term potentiation (LTP) was enhanced. However, in mice irradiated with 0.25 Gy contextual fear conditioning was enhanced and was associated with a further enhancement of the LTP magnitude. Such increase in synaptic plasticity was not seen in cognitively tested mice irradiated with 1 Gy. Thus, low dose 28Si radiation has effects on synaptic plasticity in the CA1 region of the hippocampus and these effects are modulated by cognitive testing in a contextual fear-conditioning test.


Radiation Research | 2015

Whole-Body Proton Irradiation Causes Long-Term Damage to Hematopoietic Stem Cells in Mice

Jianhui Chang; Wei Feng; Yingying Wang; Yi Luo; Antiño R. Allen; Igor Koturbash; Jennifer Turner; Blair Stewart; Jacob Raber; Martin Hauer-Jensen; Daohong Zhou; Lijian Shao

Space flight poses certain health risks to astronauts, including exposure to space radiation, with protons accounting for more than 80% of deep-space radiation. Proton radiation is also now being used with increasing frequency in the clinical setting to treat cancer. For these reasons, there is an urgent need to better understand the biological effects of proton radiation on the body. Such improved understanding could also lead to more accurate assessment of the potential health risks of proton radiation, as well as the development of improved strategies to prevent and mitigate its adverse effects. Previous studies have shown that exposure to low doses of protons is detrimental to mature leukocyte populations in peripheral blood, however, the underlying mechanisms are not known. Some of these detriments may be attributable to damage to hematopoietic stem cells (HSCs) that have the ability to self-renew, proliferate and differentiate into different lineages of blood cells through hematopoietic progenitor cells (HPCs). The goal of this study was to investigate the long-term effects of low-dose proton irradiation on HSCs. We exposed C57BL/6J mice to 1.0 Gy whole-body proton irradiation (150 MeV) and then studied the effects of proton radiation on HSCs and HPCs in the bone marrow (BM) 22 weeks after the exposure. The results showed that mice exposed to 1.0 Gy whole-body proton irradiation had a significant and persistent reduction of BM HSCs compared to unirradiated controls. In contrast, no significant changes were observed in BM HPCs after proton irradiation. Furthermore, irradiated HSCs and their progeny exhibited a significant impairment in clonogenic function, as revealed by the cobblestone area-forming cell (CAFC) and colony-forming cell assays, respectively. These long-term effects of proton irradiation on HSCs may be attributable to the induction of chronic oxidative stress in HSCs, because HSCs from irradiated mice exhibited a significant increase in NADPH oxidase 4 (NOX4) mRNA expression and reactive oxygen species (ROS) production. In addition, the increased production of ROS in HSCs was associated with a significant reduction in HSC quiescence and an increase in DNA damage. These findings indicate that exposure to proton radiation can lead to long-term HSC injury, probably in part by radiation-induced oxidative stress.


Behavioural Brain Research | 2013

Effects of whole body 56Fe radiation on contextual freezing and Arc-positive cells in the dentate gyrus

Jacob Raber; Antiño R. Allen; Susanna Rosi; Sourabh Sharma; Catherine Dayger; Matthew J. Davis; John R. Fike

The space radiation environment contains high-energy charged particles such as (56)Fe, which could pose a significant hazard to hippocampal function in astronauts during and after the mission(s). The mechanisms underlying impairments in cognition are not clear but might involve alterations in the percentage of neurons in the dentate gyrus expressing the plasticity-related immediate early gene Arc. Previously, we showed effects of cranial (56)Fe irradiation on hippocampus-dependent contextual freezing and on the percentage of Arc-positive cells in the enclosed, but not free, blade. Because it is unclear whether whole body (56)Fe irradiation causes similar effects on these markers of hippocampal function, in the present study we quantified the effects of whole body (56)Fe irradiation (600MeV, 0.5 or 1Gy) on hippocampus-dependent and hippocampus-independent cognitive performance and determined whether these effects were associated with changes in Arc expression in the enclosed and free blades of the dentate gyrus. Whole body (56)Fe irradiation impacted contextual but not cued fear freezing and the percentage of Arc-positive cells in the enclosed and free blades. In mice tested for contextual freezing, there was a correlation between Arc-positive cells in the enclosed and free blades. In addition, in mice irradiated with 0.5Gy, contextual freezing in the absence of aversive stimuli correlated with the percentage of Arc-positive cells in the enclosed blade. In mice tested for cued freezing, there was no correlation between Arc-positive cells in the enclosed and free blades. In contrast, cued freezing in the presence or absence of aversive stimuli correlated with Arc-positive cells in the free blade. In addition, in mice irradiated with 1Gy cued freezing in the absence of aversive stimuli correlated with the percentage of Arc-positive neurons in the free blade. These data indicate that while whole body (56)Fe radiation affects contextual freezing and Arc-positive cells in the dentate gyrus, the enclosed blade might be more important for contextual freezing while the free blade might be more important for cued freezing.


Radiation Research | 2016

Effects of Proton and Combined Proton and 56Fe Radiation on the Hippocampus

Jacob Raber; Antiño R. Allen; Sourabh Sharma; Barrett Allen; Susanna Rosi; Reid H. J. Olsen; Matthew J. Davis; Massarra A. Eiwaz; John R. Fike; Gregory A. Nelson

The space radiation environment contains protons and 56Fe, which could pose a significant hazard to space flight crews during and after missions. The space environment involves complex radiation exposures, thus, the effects of a dose of protons might be modulated by a dose of heavy-ion radiation. The brain, and particularly the hippocampus, may be susceptible to space radiation-induced changes. In this study, we first determined the dose-response effect of proton radiation (150 MeV) on hippocampus-dependent cognition 1 and 3 months after exposure. Based on those results, we subsequently exposed mice to protons alone (150 MeV, 0.1 Gy), 56Fe alone (600 MeV/n, 0.5 Gy) or combined proton and 56Fe radiations (protons first) with the two exposures separated by 24 h. At one month postirradiation, all animal groups showed novel object recognition. However, at three months postirradiation, mice exposed to either protons or combined proton and 56Fe radiations showed impaired novel object recognition, which was not observed in mice irradiated with 56Fe alone. The mechanisms in these impairments might involve inflammation. In mice irradiated with protons alone or 56Fe alone three months earlier, there was a negative correlation between a measure of novel object recognition and the number of newly born activated microglia in the dentate gyrus. Next, cytokine and chemokine levels were assessed in the hippocampus. At one month after exposure the levels of IL-12 were higher in mice exposed to combined radiations compared with sham-irradiated mice, while the levels of IFN-γ were lower in mice exposed to 56Fe radiation alone or combined radiations. In addition, IL-4 levels were lower in 56Fe-irradiated mice compared with proton-irradiated mice and TNF-α levels were lower in proton-irradiated mice than in mice receiving combined radiations. At three months after exposure, macrophage-derived chemokine (MDC) and eotaxin levels were lower in mice receiving combined radiations. The levels of MDC and eotaxin correlated and the levels of MDC, but not eotaxin, correlated with the percentage of newly born activated microglia in the blades of the dentate gyrus. Finally, hippocampal IL-6 levels were higher in mice receiving combined radiations compared with mice receiving 56Fe radiation alone. These data demonstrate the sensitivity of novel object recognition for detecting cognitive injury three months after exposure to proton radiation alone, and combined exposure to proton and 56Fe radiations, and that newly-born activated microglia and inflammation might be involved in this injury.


Journal of Radiation Research | 2014

Long-term epigenetic effects of exposure to low doses of 56Fe in the mouse lung

Etienne Nzabarushimana; Isabelle R. Miousse; Lijian Shao; Jianhui Chang; Antiño R. Allen; Jennifer Turner; Blair Stewart; Jacob Raber; Igor Koturbash

Despite significant progress, the long-term health effects of exposure to high charge (Z) and energy (E) nuclei (HZEs) and the underlying mechanisms remain poorly understood. Mouse studies show that space missions can result in pulmonary pathological states. The goal of this study was to evaluate the pro-fibrotic and pro-carcinogenic effects of exposure to low doses of heavy iron ions (56Fe) in the mouse lung. Exposure to 56Fe (600 MeV; 0.1, 0.2 and 0.4 Gy) resulted in minor pro-fibrotic changes, detected at the beginning of the fibrotic phase (22 weeks post exposure), which were exhibited as increased expression of chemokine Ccl3, and interleukin Il4. Epigenetic alterations were exhibited as global DNA hypermethylation, observed after exposure to 0.4 Gy. Cadm1, Cdh13, Cdkn1c, Mthfr and Sfrp1 were significantly hypermethylated after exposure to 0.1 Gy, while exposure to higher doses resulted in hypermethylation of Cdkn1c only. However, expression of these genes was not affected by any dose. Congruently with the observed patterns of global DNA methylation, DNA repetitive elements were hypermethylated after exposure to 0.4 Gy, with minor changes observed after exposure to lower doses. Importantly, hypermethylation of repetitive elements coincided with their transcriptional repression. The findings of this study will aid in understanding molecular determinants of pathological states associated with exposure to 56Fe, as well as serve as robust biomarkers for the delayed effects of irradiation. Further studies are clearly needed to investigate the persistence and outcomes of molecular alterations long term after exposure.


Radiation Research | 2013

Effects of Radiation Combined Injury on Hippocampal Function are Modulated in Mice Deficient in Chemokine Receptor 2 (CCR2)

Antiño R. Allen; Kirsten Eilertson; Sourabh Sharma; Danielle Schneider; Jennifer Baure; Barrett Allen; Susanna Rosi; Jacob Raber; John R. Fike

Chemokines and their receptors play a crucial role in normal brain function as well as in pathological conditions such as injury and disease-associated neuroinflammation. Chemokine receptor-2 (CCR2), which mediates the recruitment of infiltrating and resident microglia to sites of central nervous system (CNS) inflammation, is upregulated by ionizing irradiation and traumatic brain injury. Our objective was to determine if a deficiency in CCR2 and subsequent effects on brain microglia affect neurogenesis and cognitive function after radiation combined injury (RCI). CCR2 knock-out (–/–) and wild-type (WT) mice received 4 Gy of whole body 137Cs irradiation. Immediately after irradiation, unilateral traumatic brain injury was induced using a controlled cortical impact system. Forty-four days postirradiation, animals were tested for hippocampus-dependent cognitive performance in the Morris water-maze. After cognitive testing, animals were euthanized and their brains snap frozen for immunohistochemical assessment of neuroinflammation (activated microglia) and neurogenesis in the hippocampal dentate gyrus. All animals were able to locate the visible and hidden platform locations in the water maze; however, treatment effects were seen when spatial memory retention was assessed in the probe trials (no platform). In WT animals that received combined injury, a significant impairment in spatial memory retention was observed in the probe trial after the first day of hidden platform training (first probe trial). This impairment was associated with increased neurogenesis in the ipsilateral hemisphere of the dentate gyrus. In contrast, CCR2–/– mice, independent of insult showed significant memory retention in the first probe trial and there were no differences in the numbers of newly born neurons in the animals receiving irradiation, trauma or combined injury. Although the mechanisms involved are not clear, our data suggests that CCR2 deficiency can exert a protective effect preventing the impairment of cognitive function after combined injury.


Behavioural Brain Research | 2013

Effects of 56Fe radiation on hippocampal function in mice deficient in chemokine receptor 2 (CCR2)

Jacob Raber; Antiño R. Allen; Susanna Rosi; Sourabh Sharma; Catherine Dayger; Matthew J. Davis; John R. Fike

(56)Fe irradiation affects hippocampus-dependent cognition. The underlying mechanisms may involve alterations in neurogenesis, expression of the plasticity-related immediate early gene Arc, and inflammation. Chemokine receptor-2 (CCR2), which mediates the recruitment of infiltrating and resident microglia to sites of CNS inflammation, is upregulated by (56)Fe irradiation. CCR2 KO and wild-type mice were used to compare effects of (56)Fe radiation (600MeV, 0.25Gy) on hippocampal function using contextual fear conditioning involving tone shock pairing during training (+/+) and exposure to the same environment without tone shock pairings (-/-). In the -/- condition, irradiation enhanced habituation in WT mice, but not CCR2 KO mice, suggesting that a lack of CCR2 was associated with reduced cognitive performance. In the +/+ condition, irradiation reduced freezing but there was no genotype differences. There were no significant correlations between the number of Arc-positive cells in the dentate gyrus and freezing in either genotype. While measures of neurogenesis and gliogenesis appeared to be modulated by CCR2, there were no effects of genotype on the total numbers of newly born activated microglia before or after irradiation, indicating that other mechanisms are involved in the genotype-dependent radiation response.


PLOS ONE | 2012

The polyamine inhibitor alpha-difluoromethylornithine modulates hippocampus-dependent function after single and combined injuries

Susanna Rosi; Ryan Ferguson; Kelly Fishman; Antiño R. Allen; Jacob Raber; John R. Fike

Exposure to uncontrolled irradiation in a radiologic terrorism scenario, a natural disaster or a nuclear battlefield, will likely be concomitantly superimposed on other types of injury, such as trauma. In the central nervous system, radiation combined injury (RCI) involving irradiation and traumatic brain injury may have a multifaceted character. This may entail cellular and molecular changes that are associated with cognitive performance, including changes in neurogenesis and the expression of the plasticity-related immediate early gene Arc. Because traumatic stimuli initiate a characteristic early increase in polyamine metabolism, we hypothesized that treatment with the polyamine inhibitor alpha-difluoromethylornithine (DFMO) would reduce the adverse effects of single or combined injury on hippocampus structure and function. Hippocampal dependent cognitive impairments were quantified with the Morris water maze and showed that DFMO effectively reversed cognitive impairments after all injuries, particularly traumatic brain injury. Similar results were seen with respect to the expression of Arc protein, but not neurogenesis. Given that polyamines have been found to modulate inflammatory responses in the brain we also assessed the numbers of total and newly born activated microglia, and found reduced numbers of newly born cells. While the mechanisms responsible for the improvement in cognition after DFMO treatment are not yet clear, the present study provides new and compelling data regarding the potential use of DFMO as a potential countermeasure against the adverse effects of single or combined injury.

Collaboration


Dive into the Antiño R. Allen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

John R. Fike

University of California

View shared research outputs
Top Co-Authors

Avatar

Susanna Rosi

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Thomas Groves

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Jing Wang

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Barrett Allen

University of California

View shared research outputs
Top Co-Authors

Avatar

Frederico Kiffer

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Tyler C. Alexander

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Gwendolyn Carter

University of Arkansas for Medical Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge