Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 262
Filter
1.
Int J Mol Sci ; 22(10)2021 May 12.
Article in English | MEDLINE | ID: mdl-34065959

ABSTRACT

Brain tissue may be especially sensitive to electromagnetic phenomena provoking signs of neural stress in cerebral activity. Fifty-four adult female Sprague-Dawley rats underwent ELISA and immunohistochemistry testing of four relevant anatomical areas of the cerebrum to measure biomarkers indicating induction of heat shock protein 70 (HSP-70), glucocorticoid receptors (GCR) or glial fibrillary acidic protein (GFAP) after single or repeated exposure to 2.45 GHz radiation in the experimental set-up. Neither radiation regime caused tissue heating, so thermal effects can be ruled out. A progressive decrease in GCR and HSP-70 was observed after acute or repeated irradiation in the somatosensory cortex, hypothalamus and hippocampus. In the limbic cortex; however, values for both biomarkers were significantly higher after repeated exposure to irradiation when compared to control animals. GFAP values in brain tissue after irradiation were not significantly different or were even lower than those of nonirradiated animals in all brain regions studied. Our results suggest that repeated exposure to 2.45 GHz elicited GCR/HSP-70 dysregulation in the brain, triggering a state of stress that could decrease tissue anti-inflammatory action without favoring glial proliferation and make the nervous system more vulnerable.


Subject(s)
Cerebrum/metabolism , Glial Fibrillary Acidic Protein/metabolism , HSP70 Heat-Shock Proteins/metabolism , Receptors, Glucocorticoid/metabolism , Animals , Biomarkers/metabolism , Cerebrum/radiation effects , Female , Gene Expression Regulation/radiation effects , Hippocampus/metabolism , Hippocampus/radiation effects , Hypothalamus/metabolism , Hypothalamus/radiation effects , Rats , Rats, Sprague-Dawley , Somatosensory Cortex/metabolism , Somatosensory Cortex/radiation effects
2.
Radiat Oncol ; 15(1): 269, 2020 Nov 23.
Article in English | MEDLINE | ID: mdl-33228731

ABSTRACT

BACKGROUND: Whole-brain radiotherapy is a primary treatment for brain tumors and brain metastasis, but it also induces long-term undesired effects. Since cognitive impairment can occur, research on the etiology of secondary effects has focused on the hippocampus. Often overlooked, the hypothalamus controls critical homeostatic functions, some of which are also susceptible after whole-brain radiotherapy. Therefore, using whole-brain irradiation (WBI) in a rat model, we measured neurotransmitters and receptors in the hypothalamus. The prefrontal cortex and brainstem were also analyzed since they are highly connected to the hypothalamus and its regulatory processes. METHODS: Male Wistar rats were exposed to WBI with 11 Gy (Biologically Effective Dose = 72 Gy). After 1 month, we evaluated changes in gamma-aminobutyric acid (GABA), glycine, taurine, aspartate, glutamate, and glutamine in the hypothalamus, prefrontal cortex, and brainstem according to an HPLC method. Ratios of Glutamate/GABA and Glutamine/Glutamate were calculated. Through Western Blott analysis, we measured the expression of GABAa and GABAb receptors, and NR1 and NR2A subunits of NMDA receptors. Changes were analyzed comparing results with sham controls using the non-parametric Mann-Whitney U test (p < 0.05). RESULTS: WBI with 11 Gy induced significantly lower levels of GABA, glycine, taurine, aspartate, and GABAa receptor in the hypothalamus. Also, in the hypothalamus, a higher Glutamate/GABA ratio was found after irradiation. In the prefrontal cortex, WBI induced significant increases of glutamine and glutamate, Glutamine/Glutamate ratio, and increased expression of both GABAa receptor and NMDA receptor NR1 subunit. The brainstem showed no statistically significant changes after irradiation. CONCLUSION: Our findings confirm that WBI can affect rat brain regions differently and opens new avenues for study. After 1 month, WBI decreases inhibitory neurotransmitters and receptors in the hypothalamus and, conversely, increases excitatory neurotransmitters and receptors in the prefrontal cortex. Increments in Glutamate/GABA in the hypothalamus and Glutamine/Glutamate in the frontal cortex indicate a neurochemical imbalance. Found changes could be related to several reported radiotherapy secondary effects, suggesting new prospects for therapeutic targets.


Subject(s)
Cranial Irradiation , Hypothalamus/radiation effects , Neurotransmitter Agents/analysis , Prefrontal Cortex/radiation effects , Receptors, GABA/analysis , Receptors, N-Methyl-D-Aspartate/analysis , Animals , Brain Chemistry/radiation effects , Hypothalamus/chemistry , Male , Prefrontal Cortex/chemistry , Rats , Rats, Wistar
3.
Front Neural Circuits ; 14: 55, 2020.
Article in English | MEDLINE | ID: mdl-32973462

ABSTRACT

Background: Monochromatic blue light (MBL), with a wavelength between 400-490 nm, can regulate non-image-forming (NIF) functions of light in the central nervous system. The suprachiasmatic nucleus (SCN) in the brain is involved in the arousal-promoting response to blue light in mice. Animal and human studies showed that the responsiveness of the brain to visual stimuli is partly preserved under general anesthesia. Therefore, this study aimed to investigate whether MBL promotes arousal from sevoflurane anesthesia via activation of the SCN in mice. Methods: The induction and emergence time of sevoflurane anesthesia under MBL (460 nm and 800 lux) exposure was measured. Cortical electroencephalograms (EEGs) were recorded and the burst-suppression ratio (BSR) was calculated under MBL during sevoflurane anesthesia. The EEGs and local field potential (LFP) recordings with or without locally electrolytic ablated bilateral SCN were used to further explore the role of SCN in the arousal-promoting effect of MBL under sevoflurane anesthesia. Immunofluorescent staining of c-Fos was conducted to reveal the possible downstream mechanism of SCN activation. Results: Unlike the lack of effect on the induction time, MBL shortened the emergence time and the EEG recordings showed cortical arousal during the recovery period. MBL resulted in a significant decrease in BSR and a marked increase in EEG power at all frequency bands except for the spindle band during 2.5% sevoflurane anesthesia. MBL exposure under sevoflurane anesthesia enhances the neuronal activity of the SCN. These responses to MBL were abolished in SCN lesioned (SCNx) mice. MBL evoked a high level of c-Fos expression in the prefrontal cortex (PFC) and lateral hypothalamus (LH) compared to polychromatic white light (PWL) under sevoflurane anesthesia, while it exerted no effect on c-Fos expression in the ventrolateral preoptic area (VLPO) and locus coeruleus (LC) c-Fos expression. Conclusions: MBL promotes behavioral and electroencephalographic arousal from sevoflurane anesthesia via the activation of the SCN and its associated downstream wake-related nuclei. The clinical implications of this study warrant further study.


Subject(s)
Anesthetics, Inhalation/pharmacology , Arousal/radiation effects , Hypothalamus/radiation effects , Light , Neurons/radiation effects , Prefrontal Cortex/radiation effects , Sevoflurane/pharmacology , Suprachiasmatic Nucleus/radiation effects , Anesthesia , Animals , Brain/drug effects , Brain/metabolism , Brain/radiation effects , Electroencephalography , Hypothalamus/drug effects , Hypothalamus/metabolism , Mice , Neurons/drug effects , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-fos/radiation effects , Reflex, Righting/drug effects , Reflex, Righting/radiation effects , Suprachiasmatic Nucleus/cytology , Suprachiasmatic Nucleus/drug effects , Suprachiasmatic Nucleus/metabolism
4.
Anticancer Res ; 40(10): 5787-5792, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32988906

ABSTRACT

BACKGROUND/AIM: Hypothalamic-pituitary (HT-P) dysfunction is one of the most common endocrine late effects following cranial radiotherapy. However, there are currently no specific data describing this complication in adult-onset cancer patients after whole brain radiotherapy (WBRT). The present cohort study aims to establish the prevalence of HT-P axis dysfunction in this group of patients. PATIENTS AND METHODS: Twenty-six cancer patients previously treated with WBRT (median follow-up=20.5 months) received standardized endocrine check-up focusing on HT-P function. RESULTS: In 50% of the patients, impaired hypothalamic-pituitary function was detected during follow-up. While functional loss of a single hormonal axis was evident in 34.6% of patients, 7.7% showed an impairment of multiple endocrine axes, and one patient developed adrenocorticotropic hormone deficiency. Hypothalamic-pituitary dysfunction did not directly correlate with the applied WBRT total doses. CONCLUSION: In our cohort, hypothalamic-pituitary dysfunction appeared to be common after WBRT and was diagnosed as early as 6 months following radiation. This finding highlights the need for routine endocrine follow-up even in patients with limited life expectancy.


Subject(s)
Brain Neoplasms/radiotherapy , Cranial Irradiation/adverse effects , Hypothalamo-Hypophyseal System/radiation effects , Pituitary Gland/radiation effects , Adult , Aged , Aged, 80 and over , Brain Neoplasms/complications , Brain Neoplasms/pathology , Female , Humans , Hypothalamo-Hypophyseal System/physiopathology , Hypothalamus/physiopathology , Hypothalamus/radiation effects , Male , Middle Aged , Pituitary Gland/physiopathology , Radiation Injuries/physiopathology
5.
BMC Cancer ; 20(1): 610, 2020 Jun 30.
Article in English | MEDLINE | ID: mdl-32605648

ABSTRACT

BACKGROUND: Feasibility testing of a simultaneous sparing approach of hippocampus, hypothalamus and pituitary gland in patients undergoing whole-brain radiotherapy (WBRT) with and without a concomitant boost to metastatic sites. INTRODUCTION: Cognitive impairment and hormonal dysfunction are common side effects of cranial radiotherapy. A reduced dose application to the patho-physiologically involved functional brain areas, i.e. hippocampus, hypothalamus and pituitary gland, could reduce these common side effects. While hippocampal sparing is already a common practice to improve cognitive outcome, technical experience of additional combined sparing of the hypothalamus/pituitary gland (HT-P) is insufficient. METHODS: Twenty patients were included in the planning study. In 11 patients, a total dose of 36 Gy of WBRT (2 Gy per fraction) plus a simultaneous integrated boost (SIB) of 9 Gy (0.5 Gy per fraction, total dose: 45 Gy) to the brain metastases was applied. In 9 patients, prophylactic cranial irradiation (PCI) was simulated with a total dose of 30 Gy (2 Gy per fraction). In both patient cohorts, a sparing approach of the hippocampus and the HT-P area was simulated during WBRT. For all treatment plans, volumetric modulated arc therapy (VMAT) was used. Quality assurance included assessment of homogeneity, conformality and target coverage. RESULTS: The mean dose to the hippocampus and HT-P region was limited to less than 50% of the prescribed dose to the planning target volume (PTV) in all treatment plans. Dose homogeneity (HI) of the target volume was satisfying (median HI = 0.16 for WBRT+SIB and 0.1 for PCI) and target coverage (conformation number, CN) was not compromised (median CN = 0.82 for SIB and 0.86 for PCI). CONCLUSION: Simultaneous dose reduction to the hippocampus and the HT-P area did not compromise the PTV coverage in patients undergoing WBRT+SIB or PCI using VMAT. While the feasibility of the presented approach is promising, prospective neurologic, endocrine outcome and safety studies are required.


Subject(s)
Brain Neoplasms/radiotherapy , Cranial Irradiation/adverse effects , Organ Sparing Treatments/methods , Radiotherapy Planning, Computer-Assisted , Radiotherapy, Intensity-Modulated/adverse effects , Adult , Brain Neoplasms/diagnosis , Brain Neoplasms/secondary , Cranial Irradiation/methods , Dose Fractionation, Radiation , Dose-Response Relationship, Radiation , Feasibility Studies , Female , Hippocampus/diagnostic imaging , Hippocampus/radiation effects , Humans , Hypothalamus/diagnostic imaging , Hypothalamus/radiation effects , Male , Organ Sparing Treatments/adverse effects , Organs at Risk/diagnostic imaging , Organs at Risk/radiation effects , Pituitary Gland/diagnostic imaging , Pituitary Gland/radiation effects , Radiotherapy Dosage , Radiotherapy, Intensity-Modulated/methods , Tomography, X-Ray Computed
6.
Mol Cell Endocrinol ; 512: 110854, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32422399

ABSTRACT

Many seasonally-breeding species use daylength to time reproduction. Light-induced release of progonadal hormones involves a complex cascade of responses both inside and outside the brain. In this study, we used induction of early growth response 1 (Egr-1), the protein product of an immediate early gene, to evaluate the time course of such responses in male white-throated sparrows (Zonotrichia albicollis) exposed to a single long day. Induction of Egr-1 in the pars tuberalis began ∼11 h after dawn. This response was followed ∼6 h later by dramatic induction in the tuberal hypothalamus, including in the ependymal cells lining the third ventricle. At approximately the same time, Egr-1 was induced in dopaminergic and vasoactive intestinal peptide neurons in the tuberal hypothalamus and in dopaminergic neurons of the premammillary nucleus. We noted no induction in gonadotropin-releasing hormone (GnRH) neurons until 2 h after dawn the following morning. Overall, our results indicate that Egr-1 responses in GnRH neurons occur rather late during photostimulation, compared with responses in other cell populations, and that such induction may reflect new synthesis related to GnRH depletion rather than stimulation by light cues.


Subject(s)
Early Growth Response Protein 1/metabolism , Hypothalamus/metabolism , Photoperiod , Sexual Behavior, Animal , Sparrows/metabolism , Animals , Gene Expression Regulation/radiation effects , Hypothalamus/radiation effects , Light , Male , Seasons , Sexual Behavior, Animal/radiation effects , Songbirds/metabolism , Sparrows/physiology , Time Factors , Tissue Distribution/radiation effects
7.
Health Phys ; 119(3): 297-305, 2020 09.
Article in English | MEDLINE | ID: mdl-32384371

ABSTRACT

There is increasing evidence that the expression of non-coding RNA and mRNA (messenger RNA) is significantly altered following high-dose ionizing radiation (IR), and their expression may play a critical role in cellular responses to IR. However, the role of non-coding RNA and mRNA in radiation protection, especially in the nervous system, remains unknown. In this study, microarray profiles were used to determine microRNA (miRNA), long non-coding RNA (lncRNA), and mRNA expression in the hypothalamus of mice that were pretreated with amifostine and subsequently exposed to high-dose IR. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed. We found that fewer miRNAs, lncRNAs, and mRNAs were induced by amifostine pre-treatment in exposed mice, which exhibited antagonistic effects compared to IR, indicating that amifostine attenuated the IR-induced effects on RNA profiles. GO and KEGG pathway analyses showed changes in a variety of signaling pathways involved in inflammatory responses during radioprotection following amifostine pre-treatment in exposed mice. Taken together, our study revealed that amifostine treatment altered or attenuated miRNA, lncRNA, and mRNA expression in the hypothalamus of exposed mice. These data provide a resource to further elucidate the mechanisms underlying amifostine-mediated radioprotection in the hypothalamus.


Subject(s)
Amifostine/pharmacology , Cobalt Radioisotopes/adverse effects , Gamma Rays/adverse effects , Hypothalamus/radiation effects , MicroRNAs/radiation effects , RNA, Long Noncoding/radiation effects , RNA, Messenger/radiation effects , Radiation-Protective Agents/pharmacology , Transcriptome/radiation effects , Animals , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Whole-Body Irradiation/adverse effects
8.
Mol Cell Endocrinol ; 508: 110794, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32205144

ABSTRACT

We investigated gonadal effects on hypothalamic transcription of genes in sham-operated and castrated redheaded buntings photostimulated into spring and autumn migratory states. RNA-Seq results showed testes-dependent differences between spring and autumn migratory states. In particular, differentially expressed genes enriched G-protein-coupled receptor and calcium-ion signaling pathways during spring and autumn states, respectively. qPCR assay showed attenuated gabra5, ttr, thra and thrb expressions, suggesting reduced GABA and thyroid hormone effects on photo-sexual response in spring. In spring castrates, reduced npy, tac1 and nrcam and increased ank3 expression suggested testicular effects on the appetite, prolactin release and neuronal functions, whereas in autumn castrates, reduced rasgrp1, grm5 and grin1, and increased mras expression suggested testicular effects on the ras, G-protein and glutamate signaling pathways. Castration-induced reciprocal switching of pomc and pdyn expressions suggested effects on the overall homeostasis in both seasons. These results demonstrate transcriptome-wide changes, with season-dependent roles of testes in songbird migration.


Subject(s)
Animal Migration/physiology , Gene Expression Regulation , Hypothalamus/metabolism , Reproduction/genetics , Seasons , Songbirds/genetics , Songbirds/physiology , Animal Migration/radiation effects , Animals , Behavior, Animal/radiation effects , Body Weight/radiation effects , Castration , Gene Expression Regulation/radiation effects , Hypothalamus/radiation effects , Light , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproduction/radiation effects , Testosterone/blood , Transcriptome/genetics , Triiodothyronine/blood
9.
BMC Cancer ; 19(1): 1213, 2019 Dec 12.
Article in English | MEDLINE | ID: mdl-31830931

ABSTRACT

BACKGROUND: Cranial radiotherapy (cRT) can induce hormonal deficiencies as a consequence of significant doses to the hypothalamic-pituitary (HP) axis. In contrast to profound endocrinological follow-up data from survivors of childhood cancer treated with cRT, little knowledge exists for adult cancer patients. METHODS: A systematic search of the literature was conducted using the PubMed database and the Cochrane library offering the basis for our debate of the relevance of HP axis impairment after cRT in adult cancer patients. Against the background of potential relevance for patients receiving whole brain radiotherapy (WBRT), a particular focus was set on the temporal onset of hypopituitarism and the radiation dose to the HP axis. RESULTS: Twenty-eight original papers with a total of 1728 patients met the inclusion criteria. Radiation doses to the HP area ranged from 4 to 97 Gray (Gy). Hypopituitarism incidences ranged from 20 to 93% for adult patients with nasopharyngeal cancer or non-pituitary brain tumors. No study focused particularly on hypopituitarism after WBRT. The onset of hypopituitarism occurred as early as within the first year following cRT (range: 3 months to 25.6 years). However, since most studies started follow-up evaluation only several years after cRT, early onset of hypopituitarism might have gone unnoticed. CONCLUSION: Hypopituitarism occurs frequently after cRT in adult cancer patients. Despite the general conception that it develops only after several years, onset of endocrine sequelae can occur within the first year after cRT without a clear threshold. This finding is worth debating particularly in respect of treatment options for patients with brain metastases and favorable survival prognoses.


Subject(s)
Brain Neoplasms/radiotherapy , Cranial Irradiation/adverse effects , Hypopituitarism/etiology , Hypothalamus/radiation effects , Pituitary Gland/radiation effects , Radiation Injuries/etiology , Humans , Hypopituitarism/pathology , Hypothalamus/pathology , Pituitary Gland/pathology , Radiation Injuries/pathology , Randomized Controlled Trials as Topic
10.
Int J Biol Sci ; 15(9): 1816-1834, 2019.
Article in English | MEDLINE | ID: mdl-31523185

ABSTRACT

Disruption of the circadian rhythm is a risk factor for cancer, while glioma is a leading contributor to mortality worldwide. Substantial efforts are being undertaken to decrypt underlying molecular pathways. Our understanding of the mechanisms through which disrupted circadian rhythm induces glioma development and progression is incomplete. We, therefore, examined changes in the expression of glioma-related genes in the mouse brain after chronic jetlag (CJL) exposure. A total of 22 candidate tumor suppressor (n= 14) and oncogenes (n= 8) were identified and analyzed for their interaction with clock genes. Both the control and CJL groups were investigated for the expression of candidate genes in the nucleus accumbens, hippocampus, prefrontal cortex, hypothalamus, and striatum of wild type, Bmal1-/- and Cry1/2 double knockout male mice. We found significant variations in the expression of candidate tumor suppressor and oncogenes in the brain tissues after CJL treatment in the wild type, Bmal1-/- and Cry1/2 double knockout mice. In response to CJL treatment, some of the genes were regulated in the wild type, Bmal1-/- and Cry1/2 similarly. However, the expression of some of the genes indicated their association with the functional clock. Overall, our result suggests a link between CJL and gliomas risk at least partially dependent on the circadian clock. However, further studies are needed to investigate the molecular mechanism associated with CJL and gliomas.


Subject(s)
Brain/metabolism , Brain/pathology , Glioma/metabolism , Light , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Animals , Brain/radiation effects , Circadian Clocks/genetics , Circadian Clocks/radiation effects , Circadian Rhythm/genetics , Circadian Rhythm/radiation effects , Glioma/genetics , Hypothalamus/metabolism , Hypothalamus/pathology , Hypothalamus/radiation effects , Male , Mice , Mice, Inbred C57BL , Photoperiod , RNA, Messenger/metabolism
11.
Gen Physiol Biophys ; 38(5): 379-388, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31411574

ABSTRACT

With the rapidly increasing use of mobile phones and their close-contact usage to the brain, there are some concerns about the possible neuronal effects induced by exposure to excessive electromagnetic radiation. Exposure to a radiofrequency electromagnetic field (RF-EMF) of 835 MHz (4.0 W/kg specific absorption rate (SAR) 5 h/day for 12 weeks) may affect hypothalamic presynaptic neurons in C57BL/6 mice. The number and size of the synaptic vesicles (SVs) in the hypothalamic presynaptic terminals were significantly decreased after RF-EMF exposure. Further, the density (SVs numbers/µm) of docking and fusing SVs in the active zones of the presynaptic terminal membrane was significantly decreased in hypothalamic neurons. The expression levels of synapsin I/II and synaptotagmin 1, two regulators of SV trafficking in neurons, were also significantly decreased in the hypothalamus. In parallel, the expression of calcium channel was significantly decreased. These changes in SVs in the active zones may directly decrease the release of neurotransmitters in hypothalamic presynaptic terminals. Therefore, we further studied the possible changes in hypothalamic function by testing the core body temperature and body weight and performed the buried pellet test. The trafficking of SVs was changed by RF-EMF; however, we could not find any significant phenotypical changes in our experimental condition.


Subject(s)
Hypothalamus/metabolism , Hypothalamus/radiation effects , Radio Waves , Synaptic Vesicles/metabolism , Synaptic Vesicles/radiation effects , Animals , Biological Transport/radiation effects , Male , Mice , Mice, Inbred C57BL
12.
Cancer Med ; 8(6): 2759-2768, 2019 06.
Article in English | MEDLINE | ID: mdl-30983159

ABSTRACT

BACKGROUND: The aim of this study was to investigate dosimetric factors for predicting acute lymphopenia and the survival of glioma patients with postoperative intensity-modulated radiotherapy (IMRT). METHODS: A total of 148 glioma patients were reviewed. Acute lymphopenia was defined as a peripheral lymphocyte count (PLC) lower than 1.0 × 109 /L during radiotherapy with a normal level at pretreatment. PLCs with the corresponding dates and dose volume histogram parameters were collected. Univariate and multivariate Cox regression analyses were constructed to assess the significance of risk factors associated with lymphopenia and overall survival (OS). RESULTS: Sixty-nine (46.6%) patients developed lymphopenia during radiotherapy. Multivariate analyses revealed that the risk increased with the maximal dose of the hypothalamus (HT Dmax) ≥56 Gy (58.9% vs 28.5%, P = 0.002), minimal dose of the whole brain (WB Dmin) ≥2 Gy (54.3% vs 33.9%, P = 0.006), or mean dose of the WB (WB Dmean) ≥34 Gy (56.0% vs 37.0%, P = 0.022). Patients with older age, high-grade glioma, development of lymphopenia, high HT Dmax, WB Dmin, and WB Dmean had significantly inferior OS in the multivariate analyses. CONCLUSIONS: HT Dmax, WB Dmin, and WB Dmean are promising indicators of lymphopenia and the survival of glioma patients undergoing postoperative IMRT. The necessity and feasibility of dosimetric constraints for HT and WB is warranted with further investigation.


Subject(s)
Brain/radiation effects , Glioma/complications , Glioma/mortality , Hypothalamus/radiation effects , Lymphopenia/etiology , Lymphopenia/mortality , Radiometry , Aged , Brain Neoplasms/complications , Brain Neoplasms/diagnosis , Brain Neoplasms/mortality , Brain Neoplasms/therapy , Female , Glioma/diagnosis , Glioma/therapy , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Proportional Hazards Models , Radiotherapy/adverse effects , Radiotherapy Dosage , Retrospective Studies
13.
Annu Rev Anim Biosci ; 7: 173-194, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30332291

ABSTRACT

Organisms use changes in photoperiod for seasonal reproduction to maximize the survival of their offspring. Birds have sophisticated seasonal mechanisms and are therefore excellent models for studying these phenomena. Birds perceive light via deep-brain photoreceptors and long day-induced thyroid-stimulating hormone (TSH, thyrotropin) in the pars tuberalis of the pituitary gland (PT), which cause local thyroid hormone activation within the mediobasal hypothalamus. The local bioactive thyroid hormone controls seasonal gonadotropin-releasing hormone secretion and subsequent gonadotropin secretion. In mammals, the eyes are believed to be the only photoreceptor organ, and nocturnal melatonin secretion triggers an endocrine signal that communicates information about the photoperiod to the PT to regulate TSH. In contrast, in Salmonidae fish the input pathway to the neuroendocrine output pathway appears to be localized in the saccus vasculosus. Thus, comparative analysis is an effective way to uncover the universality and diversity of fundamental traits in various organisms.


Subject(s)
Circadian Rhythm , Reproduction/radiation effects , Vertebrates/physiology , Animals , Hypothalamus/metabolism , Hypothalamus/radiation effects , Photoperiod , Seasons , Thyroid Hormones/metabolism , Thyrotropin/metabolism
14.
Sci Rep ; 8(1): 13572, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30206280

ABSTRACT

Leptin has a central role in the maintenance of energy homeostasis, and its sensitivity is influenced by both the photoperiod and dietary polyphenols. The aim of this study was to investigate the effect of seasonal consumption of polyphenol-rich fruits on the hypothalamic leptin signaling system in non-obese and obese animals placed under different photoperiods. Non-obese and diet-induced obese male Fischer 344 rats were placed under either a short-day (SD) or long-day (LD) photoperiod and were supplemented with either 100 mg/kg of lyophilized red grapes or cherries. In non-obese animals, both fruits reduced energy balance independent of the photoperiod to which they were placed. However, the hypothalamic gene expression of Pomc was significantly up-regulated only in the SD photoperiod. In contrast, in obese animals only cherry significantly decreased the energy balance, although both fruits were able to counteract the diet-induced increase in hypothalamic AgRP mRNA levels when consumed during the SD photoperiod. In conclusion, the consumption of rich-polyphenol fruits may increase leptin sensitivity through the modulation of the hypothalamic leptin signal pathway mainly when consumed in the SD photoperiod. Therefore, fruit seasonality should be considered, as it can influence energy homeostasis and obesity.


Subject(s)
Energy Metabolism/genetics , Hypothalamus/metabolism , Leptin/metabolism , Obesity/metabolism , Polyphenols/administration & dosage , Signal Transduction , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Animals , Diet, High-Fat/adverse effects , Energy Metabolism/drug effects , Energy Metabolism/radiation effects , Freeze Drying , Fruit/chemistry , Gene Expression Regulation , Homeostasis/drug effects , Homeostasis/genetics , Homeostasis/radiation effects , Hypothalamus/drug effects , Hypothalamus/radiation effects , Leptin/genetics , Light , Male , Obesity/etiology , Obesity/genetics , Photoperiod , Proprotein Convertases/genetics , Proprotein Convertases/metabolism , Prunus avium/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Vitis/chemistry
15.
J Clin Oncol ; 36(28): 2854-2862, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30118397

ABSTRACT

PURPOSE: There are sparse data defining the dose response of radiation therapy (RT) to the hypothalamus and pituitary in pediatric and young adult patients with brain tumors. We examined the correlation between RT dose to these structures and development of endocrine dysfunction in this population. MATERIALS AND METHODS: Dosimetric and clinical data were collected from children and young adults (< 26 years of age) with brain tumors treated with proton RT on three prospective studies (2003 to 2016). Deficiencies of growth hormone (GH), thyroid hormone, adrenocorticotropic hormone, and gonadotropins were determined clinically and serologically. Incidence of deficiency was estimated using the Kaplan-Meier method. Multivariate models were constructed accounting for radiation dose and age. RESULTS: Of 222 patients in the study, 189 were evaluable by actuarial analysis, with a median follow-up of 4.4 years (range, 0.1 to 13.3 years), with 31 patients (14%) excluded from actuarial analysis for having baseline hormone deficiency and two patients (0.9%) because of lack of follow-up. One hundred thirty patients (68.8%) with medulloblastoma were treated with craniospinal irradiation (CSI) and boost; most of the remaining patients (n = 56) received involved field RT, most commonly for ependymoma (13.8%; n = 26) and low-grade glioma (7.4%; n = 14). The 4-year actuarial rate of any hormone deficiency, growth hormone, thyroid hormone, adrenocorticotropic hormone, and gonadotropin deficiencies were 48.8%, 37.4%, 20.5%, 6.9%, and 4.1%, respectively. Age at start of RT, time interval since treatment, and median dose to the combined hypothalamus and pituitary were correlated with increased incidence of deficiency. CONCLUSION: Median hypothalamic and pituitary radiation dose, younger age, and longer follow-up time were associated with increased rates of endocrinopathy in children and young adults treated with radiotherapy for brain tumors.


Subject(s)
Brain Neoplasms/radiotherapy , Cranial Irradiation/adverse effects , Hypothalamus/radiation effects , Pituitary Gland/radiation effects , Proton Therapy/adverse effects , Radiation Injuries/epidemiology , Adolescent , Child , Child, Preschool , Clinical Trials, Phase II as Topic , Cranial Irradiation/methods , Endocrine System Diseases/epidemiology , Endocrine System Diseases/etiology , Female , Humans , Male , Young Adult
16.
Proc Natl Acad Sci U S A ; 115(20): 5064-5071, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29686073

ABSTRACT

Neurotransmitter switching in the adult mammalian brain occurs following photoperiod-induced stress, but the mechanism of regulation is unknown. Here, we demonstrate that elevated activity of dopaminergic neurons in the paraventricular nucleus of the hypothalamus (PaVN) in the adult rat is required for the loss of dopamine expression after long-day photoperiod exposure. The transmitter switch occurs exclusively in PaVN dopaminergic neurons that coexpress vesicular glutamate transporter 2 (VGLUT2), is accompanied by a loss of dopamine type 2 receptors (D2Rs) on corticotrophin-releasing factor (CRF) neurons, and can lead to increased release of CRF. Suppressing activity of all PaVN glutamatergic neurons decreases the number of inhibitory PaVN dopaminergic neurons, indicating homeostatic regulation of transmitter expression in the PaVN.


Subject(s)
Brain/metabolism , Dopamine/metabolism , Dopaminergic Neurons/physiology , Light , Neurotransmitter Agents/metabolism , Stress, Physiological , Animals , Brain/pathology , Brain/radiation effects , Cells, Cultured , Corticotropin-Releasing Hormone , Dopaminergic Neurons/cytology , Hypothalamus/metabolism , Hypothalamus/pathology , Hypothalamus/radiation effects , Male , Neurotransmitter Agents/radiation effects , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/pathology , Paraventricular Hypothalamic Nucleus/radiation effects , Rats , Rats, Long-Evans , Receptors, Dopamine/metabolism , Vesicular Glutamate Transport Protein 2/metabolism
17.
Dev Neurosci ; 40(2): 120-133, 2018.
Article in English | MEDLINE | ID: mdl-29635235

ABSTRACT

Cranial radiotherapy is one of the most effective tools for treating children with brain tumors. However, radiotherapy-induced late-onset side effects have a significant impact on patients' quality of life. The purpose of this study was to investigate the effects of irradiation on metabolism and the possible molecular and cellular mechanisms behind such effects. Female Wistar rats were subjected to a single dose of 6-Gy whole-brain irradiation on postnatal day 11. The animals were sacrificed 6 h or 20 weeks after irradiation. Cell death and proliferation, microglial activation, and inflammation were analyzed and RNA sequencing was performed. We found that irradiation led to a significantly increased body weight from 15 weeks (p < 0.05) along with white adipose tissue accumulation and adipocyte hypertrophy at 20 weeks, and these changes were accompanied by glucose and lipid metabolic disturbances as indicated by reduced glucose tolerance, increased insulin resistance, increased serum triglycerides, and an increased leptin/adiponectin ratio. Furthermore, irradiation induced cell death, microglial activation, inflammation, and persistent astrocyte reactivity in the hypothalamus. Hypothalamic transcriptome analysis showed that 865 genes were downregulated and 290 genes were upregulated in the irradiated group 20 weeks after irradiation, and further pathway analysis showed that the insulin resistance-related PI3K-Akt signaling pathway and the energy expenditure-related adipocytokine signaling pathway were downregulated. Gene Ontology enrichment analysis showed that the expression of fatty acid metabolism-related proteins and effector proteins was significantly different in the irradiation group. This study demonstrates that ionizing radiation to the juvenile female brain induces hypothalamic damage that is likely to be associated with delayed metabolic abnormalities, and this critical vulnerability of the hypothalamus to irradiation should be taken into consideration in the development of future protective strategies for radiotherapy.


Subject(s)
Cranial Irradiation/adverse effects , Hypothalamus/radiation effects , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology , Animals , Female , Rats , Rats, Wistar
18.
J BUON ; 22(5): 1240-1245, 2017.
Article in English | MEDLINE | ID: mdl-29135108

ABSTRACT

PURPOSE: Hypothalamic-pituitary axis is susceptible to radiotherapy, causing endocrine disorders to childhood cancer survivors. We conducted a systematic review in order to assess the radiation-induced toxicity that leads to hormone secretion abnormalities and their severity in children with brain tumors. METHODS: The data were collected by relevant studies on PubMed and EMBASE. Articles up to December 2016 were included. We selected studies which focused on children patients (<18 yr old) with brain tumors treated with radiotherapy and the consequences for their endocrine system. RESULTS: Growth hormone (GH) deficiency was the most common post-irradiation abnormality among children cancer survivors, followed by gonadotrophin (GT), thyroid stimulating hormone (TSH), corticotropin (ACTH) and prolactin (PRL) disorders. CONCLUSIONS: The age of the patient, total radiotherapy dose, number of fractions, fraction size and the duration of treatment seem to determine the severity of these disturbances.


Subject(s)
Brain Neoplasms/complications , Hypothalamus/radiation effects , Pituitary Gland/radiation effects , Radiation Injuries/etiology , Adolescent , Brain Neoplasms/radiotherapy , Child , Child, Preschool , Female , Humans , Hypothalamus/pathology , Male , Pituitary Gland/pathology , Radiation Injuries/pathology
19.
Acta Histochem ; 119(7): 733-739, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28943001

ABSTRACT

To study the mechanism of the effect of monochromatic light on physiological function in chicken, a total of 192 newly hatched chicks were randomly divided into intact, sham-operated and pinealectomy groups then exposed to white light (WL), red light (RL), green light (GL) and blue light (BL) using a light-emitting diode (LED) system for two weeks. At P14, the hypothalami were immediately collected for immunohistochemical staining of melatonin receptor subtypes (Mel1a and Mel1b) and detection of Mel1a and Mel1b expressions using RT-PCR and western blot. Immunohistochemical staining of the hypothalamus showed that the Mel1a-ir cells were distributed in the preoptic area (POA), nucleus preopticus periventricularis (POP) and suprachiasmatic nuclei (SCN), and the Mel1b-ir cells were presented in the POA and SCN. Analysis of RT-PCR and western blot showed that the mRNA and protein levels of Mel1a and Mel1b in the hypothalamus of chick exposed to GL were increased by 10.7-29.3%, 9.18-35.9% and 8.97-27.3% compared to those in the chicks exposed to WL (P=0.029-0.002), RL (P=0.027-0.001) and BL (P=0.038-0.007) in the intact group, respectively. After pinealectomy, however, these parameters decreased and there were no significant differences among the WL, RL, GL and BL groups. These findings suggested that melatonin plays a critical role in GL illumination-enhanced Mel1a and Mel1b expressions in the hypothalamus of chicks.


Subject(s)
Chickens , Gene Expression/radiation effects , Hypothalamus/radiation effects , Light , Melatonin/metabolism , Receptors, Melatonin/genetics , Animals , Blotting, Western , Chromatography, Reverse-Phase , Male , Pineal Gland/surgery , Random Allocation
20.
Horm Behav ; 94: 53-60, 2017 08.
Article in English | MEDLINE | ID: mdl-28645693

ABSTRACT

Vocal communication in animals is important for ensuring reproductive success. Male mice emit song-like "ultrasonic vocalizations (USVs)" when they encounter female mice, and females show approach to the USVs. However, it is unclear whether USVs of male mice trigger female behavioral and endocrine responses in reproduction. In this study, we first investigated the relationship between the number of deliveries in breeding pairs for 4months and USVs syllables emitted from those paired males during 3min of sexual encounter with unfamiliar female mice. There was a positive correlation between these two indices, which suggests that breeding pairs in which males could emit USVs more frequently had more offspring. Further, we examined the effect of USVs of male mice on female sexual behavior. Female mice showed more approach behavior towards vocalizing males than devocalized males. Finally, to determine whether USVs of male mice could activate the neural system governing reproductive function in female mice, the activation of kisspeptin neurons, key neurons to drive gonadotropin-releasing hormone neurons in the hypothalamus, was examined using dual-label immunocytochemistry with cAMP response element-binding protein phosphorylation (pCREB). In the arcuate nucleus (Arc), the number of kisspeptin neurons expressing pCREB significantly increased after exposure to USVs of male as compared with noise exposure group. In conclusion, our results suggest that USVs of male mice promote fertility in female mice by activating both their approaching behavior and central kisspeptin neurons.


Subject(s)
Courtship , Hypothalamus , Kisspeptins/metabolism , Sexual Behavior, Animal/radiation effects , Ultrasonic Waves , Vocalization, Animal/physiology , Animals , Female , Hypothalamus/cytology , Hypothalamus/metabolism , Hypothalamus/radiation effects , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred ICR , Neurons/metabolism , Neurons/radiation effects , Sexual Behavior, Animal/physiology
SELECTION OF CITATIONS
SEARCH DETAIL