51
|
Schiefenhövel F, Immig K, Prodinger C, Bechmann I. Indications for cellular migration from the central nervous system to its draining lymph nodes in CD11c-GFP + bone-marrow chimeras following EAE. Exp Brain Res 2017; 235:2151-2166. [PMID: 28421248 DOI: 10.1007/s00221-017-4956-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Accepted: 04/07/2017] [Indexed: 12/25/2022]
Abstract
The concept as to how the brain maintains its immune privilege has initially been based on observations that it is lacking classical lymph vessels and later, the absence of dendritic cells (DC). This view has been challenged by several groups demonstrating drainage/migration of injected tracers and cells into cervical lymph nodes (CLNs) and the presence of brain antigens in CLNs in the course of various brain pathologies. Using CD11c-diphtheria toxin receptor (DTR)-green fluorescent protein (GFP) transgenic (tg) mice, we have shown the existence of CD11c+ cells, a main DC marker, within the brain parenchyma. Since injecting tracers or cells may cause barrier artefacts, we have now transplanted wild type (wt)-bone marrow (BM) to lethally irradiated CD11c-DTR-GFP tg mice to restrict the CD11c-DTR-GFP+ population to the brain and induced experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). We observed ramified GFP+ cells in the olfactory bulb, the cribriform plate, the nasal mucosa and superficial CLNs. We measured a significant increase of host gfp genomic DNA (gDNA) levels in lymph nodes (LNs) previously described as draining stations for the central nervous system (CNS). Using flow cytometry analysis, we observed an increase of the percentage of CD11c-GFP+ cells in brain parenchyma in the course of EAE which is most likely due to an up-regulation of CD11c of resident microglial cells since levels of gfp gDNA did not increase. Our data supports the hypothesis that brain-resident antigen presenting cells (APC) are capable of migrating to CNS-draining LNs to present myelin-associated epitopes.
Collapse
Affiliation(s)
- Fridtjof Schiefenhövel
- Medical Faculty, Institute of Anatomy, Leipzig University, Oststraße 25, 04317, Leipzig, Germany
| | - Kerstin Immig
- Medical Faculty, Institute of Anatomy, Leipzig University, Oststraße 25, 04317, Leipzig, Germany.
| | - Carolin Prodinger
- Medical Faculty, Institute of Anatomy, Leipzig University, Oststraße 25, 04317, Leipzig, Germany
| | - Ingo Bechmann
- Medical Faculty, Institute of Anatomy, Leipzig University, Oststraße 25, 04317, Leipzig, Germany
| |
Collapse
|
52
|
Najafi M, Shirazi A, Motevaseli E, Geraily G, Norouzi F, Heidari M, Rezapoor S. The melatonin immunomodulatory actions in radiotherapy. Biophys Rev 2017; 9:139-148. [PMID: 28510090 PMCID: PMC5425818 DOI: 10.1007/s12551-017-0256-8] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 03/05/2017] [Indexed: 02/07/2023] Open
Abstract
Radiotherapy has a key role in cancer treatment in more than half of patients with cancer. The management of severe side effects of this treatment modality is a limiting factor to appropriate treatment. Immune system responses play a pivotal role in many of the early and late side effects of radiation. Moreover, immune cells have a significant role in tumor response to radiotherapy, such as angiogenesis and tumor growth. Melatonin as a potent antioxidant has shown appropriate immune regulatory properties that may ameliorate toxicity induced by radiation in various organs. These effects are mediated through various modulatory effects of melatonin in different levels of tissue reaction to ionizing radiation. The effects on the DNA repair system, antioxidant enzymes, immune cells, cytokines secretion, transcription factors, and protein kinases are most important. Moreover, anti-cancer properties of melatonin may increase the therapeutic ratio of radiotherapy. Clinical applications of this agent for the management of malignancies such as breast cancer have shown promising results. It seems anti-proliferative, anti-angiogenesis, and stimulation or suppression of some immune cell responses are the main anti-tumor effects of melatonin that may help to improve response of the tumor to radiotherapy. In this review, the effects of melatonin on the modulation of immune responses in both normal and tumor tissues will be discussed.
Collapse
Affiliation(s)
- M Najafi
- Department of Medical Physics and Biomedical Engineering, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - A Shirazi
- Department of Medical Physics and Biomedical Engineering, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - E Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Gh Geraily
- Department of Medical Physics and Biomedical Engineering, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - F Norouzi
- Department of Medical Radiation Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - M Heidari
- Department of Radiology, Faculty of Paramedical, Tehran University of Medical Sciences, Tehran, Iran
| | - S Rezapoor
- Department of Radiology, Faculty of Paramedical, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
53
|
The vascular adventitia: An endogenous, omnipresent source of stem cells in the body. Pharmacol Ther 2017; 171:13-29. [DOI: 10.1016/j.pharmthera.2016.07.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 07/12/2016] [Indexed: 12/22/2022]
|
54
|
Mangoni M, Sottili M, Gerini C, Desideri I, Bastida C, Pallotta S, Castiglione F, Bonomo P, Meattini I, Greto D, Cappelli S, Di Brina L, Loi M, Biti G, Livi L. A PPAR-gamma agonist protects from radiation-induced intestinal toxicity. United European Gastroenterol J 2017; 5:218-226. [PMID: 28344789 PMCID: PMC5349355 DOI: 10.1177/2050640616640443] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 02/24/2016] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Because of its anti-inflammatory, anti-fibrotic, anti-apoptotic and anti-neoplastic properties, the PPAR-γ agonist rosiglitazone is an interesting drug for investigating for use in the prevention and treatment of radiation-induced intestinal damage. We aimed to evaluate the radioprotective effect of rosiglitazone in a murine model of acute intestinal damage, assessing whether radioprotection is selective for normal tissues or also occurs in tumour cells. METHODS Mice were total-body irradiated (12 Gy), with or without rosiglitazone (5 mg/kg/day). After 24 and 72 hours, mice were sacrificed and the jejunum was collected. HT-29 human colon cancer cells were irradiated with a single dose of 2 (1000 cells), 4 (1500 cells) or 6 (2000 cells) Gy, with or without adding rosiglitazone (20 µM) 1 hour before irradiation. HT-29-xenografted CD1 mice were irradiated (16 Gy) with or without rosiglitazone; tumour volumes were measured for 33 days. RESULTS Rosiglitazone markedly reduced histological signs of altered bowel structures, that is, villi shortening, submucosal thickening, necrotic changes in crypts, oedema, apoptosis, and inflammatory infiltrate induced by irradiation. Rosiglitazone significantly decreased p-NF-kB p65 phosphorylation and TGFβ protein expression at 24 and 72 hours post-irradiation and significantly decreased gene expression of Collagen1, Mmp13, Tnfα and Bax at 24 hours and p53 at 72 hours post-irradiation. Rosiglitazone reduced HT-29 clonogenic survival, but only produced a slight reduction of xenograft tumour growth. CONCLUSION Rosiglitazone exerts a protective effect on normal tissues and reduces alterations in bowel structures and inflammation in a radiation-induced bowel toxicity model, without interfering with the radiation effect on HT-29 cancer cells. PPAR-γ agonists should be further investigated for their application in abdominal and pelvic irradiation.
Collapse
Affiliation(s)
- Monica Mangoni
- Radiotherapy Unit, Department of
Experimental and Clinical Biomedical Sciences, University of Florence, Firenze,
Italy
| | - Mariangela Sottili
- Radiotherapy Unit, Department of
Experimental and Clinical Biomedical Sciences, University of Florence, Firenze,
Italy
| | - Chiara Gerini
- Radiotherapy Unit, Department of
Experimental and Clinical Biomedical Sciences, University of Florence, Firenze,
Italy
| | - Isacco Desideri
- Radiotherapy Unit, Department of
Experimental and Clinical Biomedical Sciences, University of Florence, Firenze,
Italy
| | - Cinzia Bastida
- Radiotherapy Unit, Department of
Experimental and Clinical Biomedical Sciences, University of Florence, Firenze,
Italy
| | - Stefania Pallotta
- Medical Physic Unit, Department of
Experimental and Clinical Biomedical Sciences, University of Florence, Firenze,
Italy
| | - Francesca Castiglione
- Department of Clinical and Experimental
Medicine, University of Florence, Firenze, Italy
| | - Pierluigi Bonomo
- Radiotherapy Unit, Department of
Experimental and Clinical Biomedical Sciences, University of Florence, Firenze,
Italy
| | - Icro Meattini
- Radiotherapy Unit, Department of
Experimental and Clinical Biomedical Sciences, University of Florence, Firenze,
Italy
| | - Daniela Greto
- Radiotherapy Unit, Department of
Experimental and Clinical Biomedical Sciences, University of Florence, Firenze,
Italy
| | - Sabrina Cappelli
- Radiotherapy Unit, Department of
Experimental and Clinical Biomedical Sciences, University of Florence, Firenze,
Italy
| | - Lucia Di Brina
- Radiotherapy Unit, Department of
Experimental and Clinical Biomedical Sciences, University of Florence, Firenze,
Italy
| | - Mauro Loi
- Radiotherapy Unit, Department of
Experimental and Clinical Biomedical Sciences, University of Florence, Firenze,
Italy
| | - Giampaolo Biti
- Radiotherapy Unit, Department of
Experimental and Clinical Biomedical Sciences, University of Florence, Firenze,
Italy
| | - Lorenzo Livi
- Radiotherapy Unit, Department of
Experimental and Clinical Biomedical Sciences, University of Florence, Firenze,
Italy
| |
Collapse
|
55
|
El-Ghazaly M, Fadel N, Rashed E, El-Batal A, Kenawy S. Anti-inflammatory effect of selenium nanoparticles on the inflammation induced in irradiated rats. Can J Physiol Pharmacol 2017; 95:101-110. [DOI: 10.1139/cjpp-2016-0183] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Selenium (Se) has been reported to possess anti-inflammatory properties, but its bioavailability and toxicity are considerable limiting factors. The present study aimed to investigate the possible anti-inflammatory and analgesic effects of selenium nanoparticles (Nano-Se) on inflammation induced in irradiated rats. Paw volume and nociceptive threshold were measured in carrageenan-induced paw edema and hyperalgesia model. Leukocytic count, tumor necrosis factor-α (TNF-α), prostaglandin E2 (PGE2), thiobarbituric acid reactive substances (TBAR), and total nitrate/nitrite (NOx) were estimated in the exudate collected from 6 day old air pouch model. Irradiated rats were exposed to 6 Gy gamma (γ)-irradiation. Nano-Se were administered orally in a dose of 2.55 mg/kg once before carrageenan injection in the first model and twice in the second model. The paw volume but not the nociceptive response produced by carrageenan in irradiated rats was higher than that induced in non-irradiated rats. Nano-Se were effective in reducing the paw volume in non-irradiated and irradiated rats but it did not alter the nociceptive threshold. The inflammation induced in irradiated rats increased all the estimated parameters in the exudate whereas; Nano-Se decreased their elevation in non-irradiated and irradiated rats. Nano-Se possess a potential anti-inflammatory activity on inflammation induced in irradiated rats.
Collapse
Affiliation(s)
- M.A. El-Ghazaly
- Drug Radiation Research Department, National Centre for Radiation Research & Technology, Atomic Energy Authority, Ahmed El-Zomor Street, Nasr City, Cairo, Egypt
| | - N. Fadel
- Drug Radiation Research Department, National Centre for Radiation Research & Technology, Atomic Energy Authority, Ahmed El-Zomor Street, Nasr City, Cairo, Egypt
| | - E. Rashed
- Drug Radiation Research Department, National Centre for Radiation Research & Technology, Atomic Energy Authority, Ahmed El-Zomor Street, Nasr City, Cairo, Egypt
| | - A. El-Batal
- Drug Radiation Research Department, National Centre for Radiation Research & Technology, Atomic Energy Authority, Ahmed El-Zomor Street, Nasr City, Cairo, Egypt
| | - S.A. Kenawy
- Pharmacology Department, Faculty of Pharmacy, Cairo University, Egypt
| |
Collapse
|
56
|
Kim S, Choe JH, Lee GJ, Kim YS, Kim SY, Lee HM, Jin HS, Kim TS, Kim JM, Cho MJ, Shin EC, Jo EK, Kim JS. Ionizing Radiation Induces Innate Immune Responses in Macrophages by Generation of Mitochondrial Reactive Oxygen Species. Radiat Res 2016; 187:32-41. [PMID: 28001907 DOI: 10.1667/rr14346.1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
During radiotherapy for tumors, the innate immune system also responds to ionizing radiation and induces immune modulation. However, little is known about the molecular mechanisms by which radiation modulates innate immune responses. In this study, we observed that radiation triggered the generation of mitochondrial reactive oxygen species (mROS), leading to innate immune responses in murine bone marrow-derived macrophages (BMDM). Radiation-induced mROS was essential for robust induction of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and IL-12p40 mRNA and protein in BMDM. Exposure to radiation also led to rapid activation of the mitogen-activated protein kinase (MAPK) and nuclear factor (NF)-κB pathways in BMDM. Notably, radiation-induced MAPK activation and NF-κB signaling were regulated by mROS in macrophages. Additionally, radiation-induced expression of TNF-α, IL-6 and IL-12p40 was dependent on JNK, p38 and NF-κB activation in BMDM. These data suggest a key role for radiation-induced pro-inflammatory responses and activation of the MAPK and NF-κB pathways through a triggering mechanism involving mROS generation.
Collapse
Affiliation(s)
- Sup Kim
- Department of aRadiation Oncology, Daejeon, Korea.,b Department of Medical Science, Daejeon, Korea.,c Department of Infection Signaling Network Research Center, Daejeon, Korea
| | - Jin Ho Choe
- b Department of Medical Science, Daejeon, Korea.,c Department of Infection Signaling Network Research Center, Daejeon, Korea
| | - Gippeum Joy Lee
- b Department of Medical Science, Daejeon, Korea.,c Department of Infection Signaling Network Research Center, Daejeon, Korea
| | - Yi Sak Kim
- b Department of Medical Science, Daejeon, Korea.,c Department of Infection Signaling Network Research Center, Daejeon, Korea
| | - Soo Yeon Kim
- b Department of Medical Science, Daejeon, Korea.,c Department of Infection Signaling Network Research Center, Daejeon, Korea
| | - Hye-Mi Lee
- b Department of Medical Science, Daejeon, Korea.,c Department of Infection Signaling Network Research Center, Daejeon, Korea
| | - Hyo Sun Jin
- b Department of Medical Science, Daejeon, Korea.,c Department of Infection Signaling Network Research Center, Daejeon, Korea
| | - Tae Sung Kim
- b Department of Medical Science, Daejeon, Korea.,c Department of Infection Signaling Network Research Center, Daejeon, Korea
| | - Jin-Man Kim
- d Department of Pathology, Chungnam National University School of Medicine, Daejeon, Korea
| | | | - Eui-Cheol Shin
- e Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, KAIST, Daejeon, Korea
| | - Eun-Kyeong Jo
- b Department of Medical Science, Daejeon, Korea.,c Department of Infection Signaling Network Research Center, Daejeon, Korea
| | - Jun-Sang Kim
- Department of aRadiation Oncology, Daejeon, Korea
| |
Collapse
|
57
|
Liu Z, Tian H, Jiang J, Yang Y, Tan S, Lin X, Liu H, Wu B. β-Arrestin-2 modulates radiation-induced intestinal crypt progenitor/stem cell injury. Cell Death Differ 2016; 23:1529-41. [PMID: 27128598 PMCID: PMC5072429 DOI: 10.1038/cdd.2016.38] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 03/14/2016] [Accepted: 03/21/2016] [Indexed: 12/24/2022] Open
Abstract
Intestinal crypt progenitor/stem (ICPS) cell apoptosis and vascular endothelial cell apoptosis are responsible for the initiation and development of ionizing radiation (IR)-evoked gastrointestinal syndrome. The signaling mechanisms underlying IR-induced ICPS cell apoptosis remain largely unclear. Our findings provide evidence that β-arrestin-2 (βarr2)-mediated ICPS cell apoptosis is crucial for IR-stimulated intestinal injury. βArr2-deficient mice exhibited decreased ICPS cell and intestinal Lgr5+ (leucine-rich repeat-containing G-protein-coupled receptor 5-positive) stem cell apoptosis, promoted crypt proliferation and reproduction, and protracted survival following lethal doses of radiation. Radioprotection in the ICPS cells isolated from βarr2-deficient mice depended on prolonged nuclear factor-κB (NF-κB) activation via direct interaction of βarr2 with IκBα and subsequent inhibition of p53-upregulated modulator of apoptosis (PUMA)-mediated mitochondrial dysfunction. Unexpectedly, βarr2 deficiency had little effect on IR-induced intestinal vascular endothelial cell apoptosis in mice. Consistently, βarr2 knockdown also provided significant radioresistance by manipulating NF-κB/PUMA signaling in Lgr5+ cells in vitro. Collectively, these observations show that targeting the βarr2/NF-κB/PUMA novel pathway is a potential radiomitigator for limiting the damaging effect of radiotherapy on the gastrointestinal system. Significance statement: acute injury to the intestinal mucosa is a major dose-limiting complication of abdominal radiotherapy. The issue of whether the critical factor for the initiation of radiation-induced intestinal injury is intestinal stem cell apoptosis or endothelial cell apoptosis remains unresolved. βArrs have recently been found to be multifunctional adaptor of apoptosis. Here, we found that β-arrestin-2 (βarr2) deficiency was associated with decreased radiation-induced ICPS cell apoptosis, which prolonged survival in abdominally irradiated mice. Moreover, βarr2 deficiency-mediated intestinal progenitor/stem cell radioprotection relied on protracted NF-κB activation and subsequent suppression of PUMA induction. Our results suggest that ICPS cell apoptosis is the factor involved in the initiation and development of radiation-induced gastrointestinal syndrome. βArr2 is a potential target for lessening radiation-induced ICPS cell apoptosis.
Collapse
Affiliation(s)
- Z Liu
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - H Tian
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - J Jiang
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Y Yang
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - S Tan
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - X Lin
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - H Liu
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - B Wu
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
58
|
Hepatoprotective effect of grape seed oil against carbon tetrachloride induced oxidative stress in liver of γ-irradiated rat. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2016; 160:1-10. [PMID: 27085796 DOI: 10.1016/j.jphotobiol.2016.03.027] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 03/21/2016] [Indexed: 12/15/2022]
Abstract
Carbon tetrachloride (CCl4) and ionizing radiation are well known environmental pollutants that generate free radicals and induce oxidative stress. The liver is the primary and major target organ responsible for the metabolism of drugs, toxic chemicals and affected by irradiation. This study investigated the effect of grape seed oil (GSO) on acute liver injury induced by carbon tetrachloride (CCl4) in γ-irradiated rats (7Gy). CCl4-intoxicated rats exhibited an elevation of ALT, AST activities, IL-6 and TNF-α level in the serum. Further, the levels of MDA, NO, NF-κB and the gene expression of CYP2E1, iNOS and Caspase-3 were increased, and SOD, CAT, GSH-Px, GST activities and GSH content were decreased. Furthermore, silent information regulator protein 1 (SIRT1) gene expression was markedly down-regulated. Additionally, alterations of the trace elements; copper, manganese, zinc and DNA fragmentation was observed in the hepatic tissues of the intoxicated group. These effects were augmented in CCl4-intoxicated-γ-irradiated rats. However, the administration of GSO ameliorated these parameters. GSO exhibit protective effects on CCl4 induced acute liver injury in γ-irradiated rats that could be attributed to its potent antioxidant, anti-inflammatory and anti-apoptotic activities. The induction of the antioxidant enzymes activities, down-regulation of the CYP2E1, iNOS, Caspase-3 and NF-κB expression, up-regulation of the trace elements concentration levels and activation of SIRT1 gene expression are responsible for the improvement of the antioxidant and anti-inflammatory status in the hepatic tissues and could be claimed to be the hepatoprotective mechanism of GSO.
Collapse
|
59
|
Huang Z, Epperly M, Watkins SC, Greenberger JS, Kagan VE, Bayır H. Necrostatin-1 rescues mice from lethal irradiation. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1862:850-856. [PMID: 26802452 PMCID: PMC4788560 DOI: 10.1016/j.bbadis.2016.01.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 11/30/2015] [Accepted: 01/19/2016] [Indexed: 12/21/2022]
Abstract
There is an emerging need in new medical products that can mitigate and/or treat the short- and long-term consequences of radiation exposure after a radiological or nuclear terroristic event. The direct effects of ionizing radiation are realized primarily via apoptotic death pathways in rapidly proliferating cells within the initial 1-2days after the exposure. However later in the course of the radiation disease necrotic cell death may ensue via direct and indirect pathways from increased generation of pro-inflammatory cytokines. Here we evaluated radiomitigative potential of necrostatin-1 after total body irradiation (TBI) and the contribution of necroptosis to cell death induced by radiation. Circulating TNFα levels were increased starting on d1 after TBI and associated with increased plasmalemma permeability in ileum of irradiated mice. Necrostatin-1 given iv. 48h after 9.5Gy TBI attenuated radiation-induced receptor interacting protein kinase 3 (RIPK3) serine phosphorylation in ileum and improved survival vs. vehicle. Utilizing apoptosis resistant cytochrome c(-/-) cells, we showed that radiation can induce necroptosis, which is attenuated by RNAi knock down of RIPK1 and RIPK3 or by treatment with necrostatin-1 or -1s whereas 1-methyl-L-tryptophan, an indoleamine-2,3-dioxygenase inhibitor, did not exhibit radiomitigative effect. This suggests that the beneficial effect of necrostatin-1 is likely through inhibition of RIPK1-mediated necroptotic pathway. Overall, our data indicate that necroptosis, a form of programmed necrosis, may play a significant role in cell death contributing to radiation disease and mortality. This study provides a proof of principle that necrostatin-1 and perhaps other RIPK1 inhibitors are promising therapeutic agents for radiomitigation after TBI.
Collapse
Affiliation(s)
- Zhentai Huang
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health, University of Pittsburgh, United States
| | - Michael Epperly
- Department of Radiation Oncology, University of Pittsburgh, United States
| | - Simon C Watkins
- Center for Biologic Imaging, Department of Cell Biology, University of Pittsburgh, United States
| | - Joel S Greenberger
- Department of Radiation Oncology, University of Pittsburgh, United States
| | - Valerian E Kagan
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health, University of Pittsburgh, United States
| | - Hülya Bayır
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health, University of Pittsburgh, United States; Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, United States; Children's Hospital of Pittsburgh, United States.
| |
Collapse
|
60
|
Antonisamy P, Arasu MV, Dhanasekaran M, Choi KC, Aravinthan A, Kim NS, Kang CW, Kim JH. Protective effects of trigonelline against indomethacin-induced gastric ulcer in rats and potential underlying mechanisms. Food Funct 2016; 7:398-408. [PMID: 26499137 DOI: 10.1039/c5fo00403a] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
The present study was undertaken to explore gastroprotective effects of trigonelline (TRG) and to determine the potential mechanisms involved in this action. In order to evaluate the gastroprotective efficiency of TRG, an indomethacin-induced ulcer model has been applied. Antioxidants, cytokines, adhesion markers and apoptosis levels have been analyzed for the biochemical mechanism involved in TRG activity. TRG (45 mg kg(-1)) pretreated rats significantly inhibited gastric lesions by 81.71%. Indomethacin administration raises the levels of leukotriene B4 (LTB4), lipid peroxidation and myeloperoxidase (MPO) with the significant declines of prostaglandin E2 (PGE2), superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GSH-px) levels. Conversely, TRG (45 mg kg(-1)) pretreated animals showed significant rises in PGE2 and antioxidant levels along with substantial reductions in LTB4, lipid peroxidation and MPO levels. Indomethacin-induced rats also exhibited considerable increases of pro-inflammatory cytokines including interleukin-6 (IL-6), interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), and interferon-γ (IFN-γ) levels and decreases of anti-inflammatory cytokines such as interleukin-10 (IL-10) and interleukin-4 (IL-4), but these imbalances were normalized through treatment of TRG. The protective activity of TRG against indomethacin-induced gastric ulcer has been ascribed to three important mechanisms: (1) anti-inflammatory; (2) antioxidant; (3) anti-apoptotic pathways.
Collapse
Affiliation(s)
- Paulrayer Antonisamy
- College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, 54596 79 Gobong-ro, Iksan-city, Jeollabuk-Do, Republic of Korea.
| | - Mariadhas Valan Arasu
- Department of Botany and Microbiology, Addiriyah Chair for Environmental Studies, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Muniappan Dhanasekaran
- Division of Ethnopharmacology, Entomology Research Institute, Loyola College, Chennai 600 034, Tamil Nadu, India
| | - Ki Choon Choi
- Grassland and forage division, National Institute of Animal Science, RDA, Seonghwan-Eup, Cheonan-Si, Chungnam 330-801, Republic of Korea
| | - Adithan Aravinthan
- College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, 54596 79 Gobong-ro, Iksan-city, Jeollabuk-Do, Republic of Korea.
| | - Nam Soo Kim
- College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, 54596 79 Gobong-ro, Iksan-city, Jeollabuk-Do, Republic of Korea.
| | - Chang-Won Kang
- College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, 54596 79 Gobong-ro, Iksan-city, Jeollabuk-Do, Republic of Korea.
| | - Jong-Hoon Kim
- College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, 54596 79 Gobong-ro, Iksan-city, Jeollabuk-Do, Republic of Korea.
| |
Collapse
|
61
|
Hellweg CE. The Nuclear Factor κB pathway: A link to the immune system in the radiation response. Cancer Lett 2015; 368:275-89. [DOI: 10.1016/j.canlet.2015.02.019] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 02/08/2015] [Accepted: 02/10/2015] [Indexed: 01/01/2023]
|
62
|
Khayyal MT, El-Hazek RM, El-Ghazaly MA. Propolis aqueous extract preserves functional integrity of murine intestinal mucosa after exposure to ionizing radiation. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2015; 40:901-906. [PMID: 26498266 DOI: 10.1016/j.etap.2015.09.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Revised: 09/07/2015] [Accepted: 09/08/2015] [Indexed: 06/05/2023]
Abstract
The ability of a specially prepared water propolis extract (PWE) to preserve the functional activity of the intestinal mucosa after radiation exposure was studied. PWE was given orally (650 mg/kg) to rats five days prior to irradiation by 6 Gy and continued for further two days. Rats were sacrificed 24h later, intestinal segments were examined histologically and homogenates were used to assess relevant biochemical parameters reflecting intestinal injury. Irradiation led to a rise in the histological damage score, a rise in tissue TNF-α and TBARS, and a decrease in sucrase, alkaline phosphatase, GSH and cholecystokinin as well as a decrease in plasma citrulline. The findings reflect a decrease in intestinal functional activity. PWE preserved the intestinal integrity and largely protected against the changes induced in the histology damage score and all parameters measured, possibly as a result of the antioxidant and anti-inflammatory action of its caffeic acid content.
Collapse
Affiliation(s)
- Mohamed T Khayyal
- Department of Pharmacology, Faculty of Pharmacy, Cairo University, Egypt.
| | - Rania M El-Hazek
- Department of Drug Radiation Research, National Centre for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Mona A El-Ghazaly
- Department of Drug Radiation Research, National Centre for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
63
|
Grellier N, Deray G, Yousfi A, Khodari W, Bouaita R, Belkacemi Y. Carence martiale fonctionnelle, inflammation et fatigue après radiothérapie. Bull Cancer 2015; 102:780-5. [DOI: 10.1016/j.bulcan.2015.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 12/17/2014] [Indexed: 01/20/2023]
|
64
|
Acharya MM, Patel NH, Craver BM, Tran KK, Giedzinski E, Tseng BP, Parihar VK, Limoli CL. Consequences of low dose ionizing radiation exposure on the hippocampal microenvironment. PLoS One 2015; 10:e0128316. [PMID: 26042591 PMCID: PMC4456101 DOI: 10.1371/journal.pone.0128316] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 04/24/2015] [Indexed: 01/24/2023] Open
Abstract
The response of the brain to irradiation is complex, involving a multitude of stress inducible pathways that regulate neurotransmission within a dynamic microenvironment. While significant past work has detailed the consequences of CNS radiotherapy following relatively high doses (≥ 45 Gy), few studies have been conducted at much lower doses (≤ 2 Gy), where the response of the CNS (like many other tissues) may differ substantially from that expected from linear extrapolations of high dose data. Low dose exposure could elicit radioadaptive modulation of critical CNS processes such as neurogenesis, that provide cellular input into hippocampal circuits known to impact learning and memory. Here we show that mice deficient for chemokine signaling through genetic disruption of the CCR2 receptor exhibit a neuroprotective phenotype. Compared to wild type (WT) animals, CCR2 deficiency spared reductions in hippocampal neural progenitor cell survival and stabilized neurogenesis following exposure to low dose irradiation. While radiation-induced changes in microglia levels were not found in WT or CCR2 deficient animals, the number of Iba1+ cells did differ between each genotype at the higher dosing paradigms, suggesting that blockade of this signaling axis could moderate the neuroinflammatory response. Interestingly, changes in proinflammatory gene expression were limited in WT animals, while irradiation caused significant elevations in these markers that were attenuated significantly after radioadaptive dosing paradigms in CCR2 deficient mice. These data point to the importance of chemokine signaling under low dose paradigms, findings of potential significance to those exposed to ionizing radiation under a variety of occupational and/or medical scenarios.
Collapse
Affiliation(s)
- Munjal M. Acharya
- Department of Radiation Oncology, University of California Irvine, Irvine, CA, 92697–2695, United States of America
| | - Neal H. Patel
- Department of Radiation Oncology, University of California Irvine, Irvine, CA, 92697–2695, United States of America
| | - Brianna M. Craver
- Department of Radiation Oncology, University of California Irvine, Irvine, CA, 92697–2695, United States of America
| | - Katherine K. Tran
- Department of Radiation Oncology, University of California Irvine, Irvine, CA, 92697–2695, United States of America
| | - Erich Giedzinski
- Department of Radiation Oncology, University of California Irvine, Irvine, CA, 92697–2695, United States of America
| | - Bertrand P. Tseng
- Department of Radiation Oncology, University of California Irvine, Irvine, CA, 92697–2695, United States of America
| | - Vipan K. Parihar
- Department of Radiation Oncology, University of California Irvine, Irvine, CA, 92697–2695, United States of America
| | - Charles L. Limoli
- Department of Radiation Oncology, University of California Irvine, Irvine, CA, 92697–2695, United States of America
- * E-mail:
| |
Collapse
|
65
|
Chen YX, Zeng ZC, Sun J, Zhang ZY, Zeng HY, Hu WX. Radioprotective effect of kupffer cell depletion on hepatic sinusoidal endothelial cells. Radiat Res 2015; 183:563-70. [PMID: 25897555 DOI: 10.1667/rr13869.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Radiation-induced liver injury remains a clinical problem and data suggest that sinusoidal endothelial cells (SECs) are an important target. The purpose of this study was to determine whether the inhibition of Kupffer cells before exposure would protect SECs from radiation-induced injury. Sprague-Dawley rats were intravenously injected 24 h before irradiation with Kupffer cell inhibitor gadolinium chloride (GdCl3) (10 mg/kg body weight). Three groups of animals were treated: 1. control group (saline and sham irradiation); 2. GdCl3 + 30 Gy radiation group and 3. 30 Gy radiation only group. Specimens were collected at 2, 6, 12, 24 and 48 h after completion of each treatment. Liver tissue was assessed for inflammatory cytokine expression and radiation-induced SEC injury based on serum hyaluronic acid (HA) level, apoptosis and ultrastructural and histological analyses. The results showed that radiation exposure caused apoptosis of SECs, but not hepatocytes. Inflammatory cytokine expression, including tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) expression, was significantly attenuated in the GdCl3 + 30 Gy radiation group, compared with the 30 Gy radiation-only group (P < 0.05). The GdCl3 + radiation-treated rats exhibited significantly lower levels of HA and SEC apoptosis than the radiation-treated only rats at early time points, and radiation-induced liver injury was also attenuated. In conclusion, we hypothesize that selective Kupffer cell inhibition by gadolinium chloride was shown to reduce apoptosis in SECs caused by irradiation of the live and protected the liver against radiation-induced injury.
Collapse
Affiliation(s)
- Yi-Xing Chen
- a Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | | | | | | | | | | |
Collapse
|
66
|
Chu J, Zhang X, Jin L, Chen J, Du B, Pang Q. Protective effects of caffeic acid phenethyl ester against acute radiation-induced hepatic injury in rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2015; 39:683-689. [PMID: 25704035 DOI: 10.1016/j.etap.2015.01.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 01/27/2015] [Accepted: 01/31/2015] [Indexed: 06/04/2023]
Abstract
Caffeic acid phenyl ester (CAPE) is a potent anti-inflammatory agent and it can eliminate the free radicals. The current study was intended to evaluate the protective effect of CAPE against the acute radiation-induced liver damage in rats. Male Sprague-Dawley rats were intraperitoneally administered with CAPE (30 mg/kg) for 3 consecutive days before exposing them to a single dose of 30 Gy of β-ray irradiation to upper abdomen. We found that pretreatment with CAPE significantly decreased the serum levels of alanine aminotransferase and aspartate aminotransferase and increased the activity of superoxide dismutase and glutathione. Histological evaluation further confirmed the protection of CAPE against radiation-induced hepatotoxicity. TUNEL assay showed that CAPE pretreatment inhibited hepatocyte apoptosis. Moreover, CAPE inhibited the nuclear transport of NF-κB p65 subunit, decreased the level of tumor necrosis factor-α, nitric oxide and inducible nitric oxide synthase. Taken together, these results suggest that pretreatment with CAPE offers protection against radiation-induced hepatic injury.
Collapse
Affiliation(s)
- JianJun Chu
- Department of Oncology, The Affiliated Hospital of Jiangnan University, 200 Huihe Road, 214122 Wuxi, China
| | - Xiaojun Zhang
- Department of Oncology, The Affiliated Hospital of Jiangnan University, 200 Huihe Road, 214122 Wuxi, China
| | - Liugen Jin
- Department of Oncology, The Affiliated Hospital of Jiangnan University, 200 Huihe Road, 214122 Wuxi, China
| | - Junliang Chen
- Wuxi Medical School, Jiangnan University, 1800 Lihu Road, 214122 Wuxi, China
| | - Bin Du
- Wuxi Medical School, Jiangnan University, 1800 Lihu Road, 214122 Wuxi, China
| | - Qingfeng Pang
- Wuxi Medical School, Jiangnan University, 1800 Lihu Road, 214122 Wuxi, China.
| |
Collapse
|
67
|
Elkady AA, Ibrahim IM. Protective effects of erdosteine against nephrotoxicity caused by gamma radiation in male albino rats. Hum Exp Toxicol 2015; 35:21-8. [PMID: 25716170 DOI: 10.1177/0960327115574919] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The aim of this study was focused on investigating the possible protective effect of erdosteine against gamma radiation-induced renal lesions in male albino rats. Twenty-eight albino rats were divided into four equal groups as follows: control group, irradiated group (animals subjected to whole-body gamma irradiation at a dose of 5 Gy), treated group (each rat received 100 mg/kg body weight once daily, orally by gastric tube, erdosteine for 1 week), and treated irradiated group (each rat received 100 mg/kg body weight once daily, orally by gastric tube, erdosteine for 1 week, then exposed to whole-body gamma irradiation at a dose of 5 Gy). The results revealed that the administration of erdosteine to rats before irradiation significantly ameliorated the changes occurred in kidney function (creatinine and urea) compared with irradiated group. Also the changes in serum tumor necrosis factor α, interleukin 1β, and interleukin 6 activities were markedly improved compared with the corresponding values of irradiated group. Kidney catalase and glutathione peroxidase (GPx) activities and reduced glutathione concentration showed approximately normal level when compared with the irradiated group. The histopathological results showed distinctive pattern of renal lesions in irradiated group, while in treated irradiated group the renal tissues showed relatively well-preserved architecture. Erdosteine acts in the kidney as a potent scavenger of free radicals to prevent or ameliorate the toxic effects of gamma irradiation as shown in the biochemical and histopathological changes and might provide substantial protection against radiation-induced inflammatory damage.
Collapse
Affiliation(s)
- A A Elkady
- Health Radiation Research Department, National Centre for Radiation Research and Technology, Cairo, Egypt
| | - I M Ibrahim
- Health Radiation Research Department, National Centre for Radiation Research and Technology, Cairo, Egypt
| |
Collapse
|
68
|
Di Maggio FM, Minafra L, Forte GI, Cammarata FP, Lio D, Messa C, Gilardi MC, Bravatà V. Portrait of inflammatory response to ionizing radiation treatment. J Inflamm (Lond) 2015; 12:14. [PMID: 25705130 PMCID: PMC4336767 DOI: 10.1186/s12950-015-0058-3] [Citation(s) in RCA: 196] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 01/29/2015] [Indexed: 01/05/2023] Open
Abstract
Ionizing radiation (IR) activates both pro-and anti-proliferative signal pathways producing an imbalance in cell fate decision. IR is able to regulate several genes and factors involved in cell-cycle progression, survival and/or cell death, DNA repair and inflammation modulating an intracellular radiation-dependent response. Radiation therapy can modulate anti-tumour immune responses, modifying tumour and its microenvironment. In this review, we report how IR could stimulate inflammatory factors to affect cell fate via multiple pathways, describing their roles on gene expression regulation, fibrosis and invasive processes. Understanding the complex relationship between IR, inflammation and immune responses in cancer, opens up new avenues for radiation research and therapy in order to optimize and personalize radiation therapy treatment for each patient.
Collapse
Affiliation(s)
- Federica Maria Di Maggio
- />Department of Pathobiology and Medical and Forensic Biotechnologies, University of Palermo, Palermo, Italy
- />IBFM CNR – LATO, Contrada Pietrapollastra Pisciotto, Cefalù, PA Italy
| | - Luigi Minafra
- />IBFM CNR – LATO, Contrada Pietrapollastra Pisciotto, Cefalù, PA Italy
| | - Giusi Irma Forte
- />IBFM CNR – LATO, Contrada Pietrapollastra Pisciotto, Cefalù, PA Italy
| | | | - Domenico Lio
- />Department of Pathobiology and Medical and Forensic Biotechnologies, University of Palermo, Palermo, Italy
| | - Cristina Messa
- />IBFM CNR – LATO, Contrada Pietrapollastra Pisciotto, Cefalù, PA Italy
- />Department of Health Sciences, Tecnomed Foundation, University of Milano-Bicocca, Milan, Italy
- />Nuclear Medicine Center, San Gerardo Hospital, Monza, Italy
| | - Maria Carla Gilardi
- />IBFM CNR – LATO, Contrada Pietrapollastra Pisciotto, Cefalù, PA Italy
- />Department of Health Sciences, Tecnomed Foundation, University of Milano-Bicocca, Milan, Italy
- />Nuclear Medicine, San Raffaele Scientific Institute, Milan, Italy
| | - Valentina Bravatà
- />IBFM CNR – LATO, Contrada Pietrapollastra Pisciotto, Cefalù, PA Italy
| |
Collapse
|
69
|
El-Ghazaly MA, El-Hazek RM, Khayyal MT. Protective effect of the herbal preparation, STW 5, against intestinal damage induced by gamma radiation in rats. Int J Radiat Biol 2015; 91:150-6. [DOI: 10.3109/09553002.2014.954059] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
70
|
Caffeic acid phenethyl ester and therapeutic potentials. BIOMED RESEARCH INTERNATIONAL 2014; 2014:145342. [PMID: 24971312 PMCID: PMC4058104 DOI: 10.1155/2014/145342] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 04/02/2014] [Accepted: 04/14/2014] [Indexed: 12/13/2022]
Abstract
Caffeic acid phenethyl ester (CAPE) is a bioactive compound of propolis extract. The literature search elaborates that CAPE possesses antimicrobial, antioxidant, anti-inflammatory, and cytotoxic properties. The principal objective of this review article is to sum up and critically assess the existing data about therapeutic effects of CAPE in different disorders. The findings elaborate that CAPE is a versatile therapeutically active polyphenol and an effective adjuvant of chemotherapy for enhancing therapeutic efficacy and diminishing chemotherapy-induced toxicities.
Collapse
|
71
|
Camara-Lemarroy CR. Remote ischemic preconditioning as treatment for non-ischemic gastrointestinal disorders: Beyond ischemia-reperfusion injury. World J Gastroenterol 2014; 20:3572-3581. [PMID: 24707140 PMCID: PMC3974524 DOI: 10.3748/wjg.v20.i13.3572] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 10/23/2013] [Accepted: 01/02/2014] [Indexed: 02/06/2023] Open
Abstract
Common gastrointestinal diseases such as radiation enteritis (RE), acute pancreatitis, inflammatory bowel diseases (IBD) and drug-induced hepatotoxicity share pathophysiological mechanisms at the molecular level, mostly involving the activation of many pathways of the immune response, ultimately leading to tissue injury. Increased oxidative stress, inflammatory cytokine release, inflammatory cell infiltration and activation and the up-regulation of inflammatory transcription factors participate in the pathophysiology of these complex entities. Treatment varies in each specific disease, but at least in the cases of RE and IBD immunosuppressors are effective. However, full therapeutic responses are not always achieved. The pathophysiology of ischemia-reperfusion (IR) injury shares many of these mechanisms. Brief and repetitive periods of ischemia in an organ or limb have been shown to protect against subsequent major IR injury in distant organs, a phenomenon called remote ischemic preconditioning (RIP). This procedure has been shown to protect the gut, pancreas and liver by modulating many of the same inflammatory mechanisms. Since RIP is safe and tolerable, and has shown to be effective in some recent clinical trials, I suggest that RIP could be used as a physiologically relevant adjunct treatment for non-ischemic gastrointestinal inflammatory conditions.
Collapse
|
72
|
Morganti JM, Jopson TD, Liu S, Gupta N, Rosi S. Cranial irradiation alters the brain's microenvironment and permits CCR2+ macrophage infiltration. PLoS One 2014; 9:e93650. [PMID: 24695541 PMCID: PMC3973545 DOI: 10.1371/journal.pone.0093650] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 03/08/2014] [Indexed: 12/24/2022] Open
Abstract
Therapeutic irradiation is commonly used to treat primary or metastatic central nervous system tumors. It is believed that activation of neuroinflammatory signaling pathways contributes to the development of common adverse effects, which may ultimately contribute to cognitive dysfunction. Recent studies identified the chemokine (C-C motif) receptor (CCR2), constitutively expressed by cells of the monocyte-macrophage lineage, as a mediator of cognitive impairments induced by irradiation. In the present study we utilized a unique reporter mouse (CCR2RFP/+CX3CR1GFP/+) to accurately delineate the resident (CX3CR1+) versus peripheral (CCR2+) innate immune response in the brain following cranial irradiation. Our results demonstrate that a single dose of 10Gy cranial γ-irradiation induced a significant decrease in the percentage of resident microglia, while inducing an increase in the infiltration of peripherally derived CCR2+ macrophages. Although reduced in percentage, there was a significant increase in F4/80+ activated macrophages in irradiated animals compared to sham. Moreover, we found that there were altered levels of pro-inflammatory cytokines, chemokines, adhesion molecules, and growth factors in the hippocampi of wild type irradiated mice as compared to sham. All of these molecules are implicated in the recruitment, adhesion, and migration of peripheral monocytes to injured tissue. Importantly, there were no measureable changes in the expression of multiple markers associated with blood-brain barrier integrity; implicating the infiltration of peripheral CCR2+ macrophages may be due to inflammatory induced chemotactic signaling. Cumulatively, these data provide evidence that therapeutic levels of cranial radiation are sufficient to alter the brain’s homeostatic balance and permit the influx of peripherally-derived CCR2+ macrophages as well as the regional susceptibility of the hippocampal formation to ionizing radiation.
Collapse
Affiliation(s)
- Josh M. Morganti
- Brain and Spinal Injury Center, University of California San Francisco, San Francisco, California, United States of America
- Departments of Physical Therapy and Rehabilitation Science, University of California San Francisco, San Francisco, California, United States of America
| | - Timothy D. Jopson
- Brain and Spinal Injury Center, University of California San Francisco, San Francisco, California, United States of America
- Departments of Physical Therapy and Rehabilitation Science, University of California San Francisco, San Francisco, California, United States of America
| | - Sharon Liu
- Neurological Surgery, University of California San Francisco, San Francisco, California, United States of America
| | - Nalin Gupta
- Neurological Surgery, University of California San Francisco, San Francisco, California, United States of America
| | - Susanna Rosi
- Brain and Spinal Injury Center, University of California San Francisco, San Francisco, California, United States of America
- Departments of Physical Therapy and Rehabilitation Science, University of California San Francisco, San Francisco, California, United States of America
- Neurological Surgery, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
73
|
Kma L. Plant Extracts and Plant-Derived Compounds: Promising Players in Countermeasure Strategy Against Radiological Exposure: A Review. Asian Pac J Cancer Prev 2014; 15:2405-25. [DOI: 10.7314/apjcp.2014.15.6.2405] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
74
|
Genistein Alleviates Radiation-Induced Pneumonitis by Depressing Ape1/Ref-1 Expression to Down-regulate Inflammatory Cytokines. Cell Biochem Biophys 2014; 69:725-33. [DOI: 10.1007/s12013-014-9859-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
75
|
Garg S, Wang W, Prabath BG, Boerma M, Wang J, Zhou D, Hauer-Jensen M. Bone marrow transplantation helps restore the intestinal mucosal barrier after total body irradiation in mice. Radiat Res 2014; 181:229-39. [PMID: 24568131 DOI: 10.1667/rr13548.1] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bone marrow transplantation (BMT) substantially improves 10-day survival after total body irradiation (TBI), consistent with an effect on intestinal radiation death. Total body irradiation, in addition to injuring the intestinal epithelium, also perturbs the mucosal immune system, the largest immune system in the body. This study focused on how transplanted bone marrow cells (BMCs) help restore mucosal immune cell populations after sublethal TBI (8.0 Gy). We further evaluated whether transplanted BMCs: (a) home to sites of radiation injury using green fluorescent protein labeled bone marrow; and (b) contribute to restoring the mucosal barrier in vivo. As expected, BMT accelerated recovery of peripheral blood (PB) cells. In the intestine, BMT was associated with significant early recovery of mucosal granulocytes (P = 0.005). Bone marrow transplantation did not affect mucosal macrophages or lymphocyte populations at early time points, but enhanced the recovery of these cells from day 14 onward (P = 0.03). Bone marrow transplantation also attenuated radiation-induced increase of intestinal CXCL1 and restored IL-10 levels (P = 0.001). Most importantly, BMT inhibited the post-radiation increase in intestinal permeability after 10 Gy TBI (P = 0.02) and modulated the expression of tight junction proteins (P = 0.01-0.05). Green fluorescent protein-positive leukocytes were observed both in intestinal tissue and in PB. These findings strongly suggest that BMT, in addition to enhancing general hematopoietic and immune system recovery, helps restore the intestinal immune system and enhances intestinal mucosal barrier function. These findings may be important in the development and understanding of strategies to alleviate or treat intestinal radiation toxicity.
Collapse
Affiliation(s)
- Sarita Garg
- a Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | | | | | | | | | | | | |
Collapse
|
76
|
Gupta M, Rana P, Trivedi R, Kumar BSH, Khan AR, Soni R, Rathore RKS, Khushu S. Comparative evaluation of brain neurometabolites and DTI indices following whole body and cranial irradiation: a magnetic resonance imaging and spectroscopy study. NMR IN BIOMEDICINE 2013; 26:1733-1741. [PMID: 24038203 DOI: 10.1002/nbm.3010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 06/27/2013] [Accepted: 07/15/2013] [Indexed: 06/02/2023]
Abstract
Understanding early differential response of brain during whole body radiation or cranial radiation exposure is of significant importance for better injury management during accidental or intentional exposure to ionizing radiation. We investigated the early microstructural and metabolic profiles using in vivo diffusion tensor imaging (DTI) and proton magnetic resonance spectroscopy ((1)H MRS) following whole body and cranial radiation exposure of 8 Gy in mice using a 7.0 T animal MRI system and compared profiles with sham controls at days 1, 3, 5 and 10 post irradiation. A significant decrease in fractional anisotropy (FA) values was found in hippocampus, thalamic and hypothalamic regions (p < 0.05) in both whole body and cranial irradiated groups compared with controls, suggesting radiation induced reactive astrogliosis or neuroinflammatory response. In animals exposed to whole body radiation, FA was significantly decreased in some additional brain regions such as sensory motor cortex and corpus callosum in comparison with cranial irradiation groups and controls. Changes in FA were observed till day 10 post irradiation in both the groups. However, MRS study from hippocampus revealed changes only in the whole body radiation dose group. Significant reduction in the ratios of the metabolites myoinositol (mI, p = 0.02) and taurine (tau, p = 0.03) to total creatine were observed, and these metabolic alterations persisted till day 10 post irradiation. To the best of our knowledge this study has for the first time documented a comparative account of microstructural and metabolic aspects of whole body and cranial radiation induced early brain injury using in vivo MRI. Overall our findings suggest differential response at microstructure and metabolite levels following cranial or whole body radiation exposure.
Collapse
Affiliation(s)
- Mamta Gupta
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | | | | | | | | | | | | | | |
Collapse
|
77
|
Kma L. Synergistic Effect of Resveratrol and Radiotherapy in Control of Cancers. Asian Pac J Cancer Prev 2013; 14:6197-208. [DOI: 10.7314/apjcp.2013.14.11.6197] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
78
|
Bigildeev AE, Zhironkina OA, Lubkova ON, Drize NJ. Interleukin-1 beta is an irradiation-induced stromal growth factor. Cytokine 2013; 64:131-7. [PMID: 23962752 DOI: 10.1016/j.cyto.2013.07.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 06/14/2013] [Accepted: 07/02/2013] [Indexed: 12/25/2022]
Abstract
Gamma irradiation of tissues and organs leads to many pathological consequences due to the formation of reactive oxygen species, DNA damage and the subsequent massive death of cells. The therapeutic use of gamma irradiation in the treatment of cancer is based on its penetrating power and damaging effects on tumor cells. Other effects from the irradiation are unnoticeable in comparison. Moreover, the long-term consequences of gamma irradiation are still poorly understood. When a donor bone marrow plug is implanted under the renal capsule of a syngeneic animal, а hematopoietic ectopic focus is formed. The size of the focus is increased in mice that received irradiation compared to non-irradiated ones, regardless of the amount of time between irradiation and bone marrow plug implantation. Long-term repetitive injections of blood serum from irradiated mice given to syngeneic non-irradiated recipients of bone marrow plugs also lead to the formation of enlarged foci. Hence, the blood of irradiated animals must contain an activity that induces the growth of a hematopoietic microenvironment. It was previously shown that the bones of irradiated animals secrete a growth factor required to create stromal microenvironments. The identity of this factor has, until now, been difficult to obtain. We demonstrated that interleukin 1 beta (IL-1) stimulates the growth of murine bone marrow stromal cells in vitro and in vivo. It was shown that the expression of the Il1b gene and the secretion of its product, IL-1, were activated in bone cells long after total body gamma irradiation. Hence, IL-1, or proteins regulated by this cytokine, appears to be the same stromal growth factor previously observed in the serum of irradiated animals. Our data demonstrate several non-canonical functions of IL-1. In addition, the presence of up-regulated levels of IL-1 long after irradiation points to an unknown mechanism governing its gene expression.
Collapse
Affiliation(s)
- Alexey E Bigildeev
- Laboratory Physiology of Hematopoiesis, Hematological Research Center, Ministry of Health, Noviy Zikovskiy proezd 4, 125167 Moscow, Russian Federation.
| | | | | | | |
Collapse
|
79
|
Uçan MC, Koparal M, Ağaçayak S, Gunay A, Ozgoz M, Atilgan S, Yaman F. Influence of caffeic acid phenethyl ester on bone healing in a rat model. J Int Med Res 2013; 41:1648-54. [PMID: 24065455 DOI: 10.1177/0300060513490613] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE To examine the effects of caffeic acid phenethyl ester (CAPE; a component of honey bee-hive propolis with antioxidant, anti-inflammatory, antiviral and anticancer properties) on bone regeneration and fibrotic healing in a rat model. METHODS Male Sprague-Dawley rats (n = 63; mean age 7 weeks; weight 280-490 g) were randomly divided into three groups: A, cranial defect with no bone healing treatment (n = 21); B, cranial defect treated with CAPE (n = 21); C, cranial defect treated with CAPE and β-tricalcium phosphate/hydroxyl apatite (n = 21). Rats were anaesthetized with ketamine (8 mg/100 g) by intraperitoneal injection and a cranial critical size bone defect was created. Following surgery, CAPE (10 µmol/kg) was administered by daily intraperitoneal injection. Seven rats in each group were killed at days 7, 15 and 30 following surgery. Bone regeneration, fibrotic healing and osteoblast activity were evaluated by histopathology. RESULTS Statistically significant differences in healing were found between all groups. There were no statistically significant within-group differences between day 7 and 15. At day 30, bone healing scores were significantly higher in groups B and C compared with group A. CONCLUSION CAPE significantly improved bone-defect healing in a rat model, suggesting that CAPE has beneficial effects on bone healing.
Collapse
Affiliation(s)
- M C Uçan
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Dicle University, Turkey
| | | | | | | | | | | | | |
Collapse
|
80
|
Tőkés T, Varga G, Garab D, Nagy Z, Fekete G, Tuboly E, Plangár I, Mán I, Szabó RE, Szabó Z, Volford G, Ghyczy M, Kaszaki J, Boros M, Hideghéty K. Peripheral inflammatory activation after hippocampus irradiation in the rat. Int J Radiat Biol 2013; 90:1-6. [DOI: 10.3109/09553002.2013.836617] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
81
|
Akpolat M, Gulle K, Topcu-Tarladacalisir Y, Safi Oz Z, Bakkal BH, Arasli M, Ozel Turkcu U. Protection by L-carnitine against radiation-induced ileal mucosal injury in the rat: pattern of oxidative stress, apoptosis and cytokines. Int J Radiat Biol 2013; 89:732-740. [PMID: 23510242 DOI: 10.3109/09553002.2013.787176] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE In this study, we tested the effects of L-carnitine (LC) on radiation-induced ileal mucosal damage. MATERIALS AND METHODS Thirty Wistar albino rats were divided into five groups. The control group received physiological saline intraperitoneally (i.p.). Radiation-1 and radiation-2 groups received whole-body X-irradiation of 8.3 Gy as a single dose. These groups were sacrificed at the 6th hour and 4th day after irradiation, respectively. The Radiation-1 + LC and the radiation-2 + LC groups received the same dose irradiation plus a daily dose of 200 mg/kg LC. LC was applied one day before and for four days after irradiation. RESULTS The levels of serum monocyte chemotactic protein-1 (MCP-1) and interferon gamma (IFN-γ) were significantly higher in the radiation groups when compared with the control. Treatment with LC decreased the serum MCP-1 and IFN-γ levels considerably. In the radiations groups, the Chiu score was significantly elevated compared with that of the control group. However, LC administered prior to the irradiation reduced the severity of mucosal damage. The number of apoptotic cells of the ileal crypt in the irradiated rats increased from the 6th hour after irradiation and then decreased at 4th day. CONCLUSIONS Our data demonstrated that LC may be beneficial to radiation enteritis.
Collapse
Affiliation(s)
- Meryem Akpolat
- Department of Histology and Embryology, Faculty of Medicine, Bulent Ecevit University, Zonguldak, Turkey.
| | | | | | | | | | | | | |
Collapse
|
82
|
Tolba MF, Azab SS, Khalifa AE, Abdel-Rahman SZ, Abdel-Naim AB. Caffeic acid phenethyl ester, a promising component of propolis with a plethora of biological activities: a review on its anti-inflammatory, neuroprotective, hepatoprotective, and cardioprotective effects. IUBMB Life 2013; 65:699-709. [PMID: 23847089 DOI: 10.1002/iub.1189] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 05/20/2013] [Indexed: 01/10/2023]
Abstract
Caffeic acid phenethyl ester (CAPE) is an important active component of honey bee propolis that possesses a plethora of biological activities. Propolis is used safely in traditional medicine as a dietary supplement for its therapeutic benefits. This review highlights the recently published data about CAPE bioavailability, anti-inflammatory, neuroprotective; hepatoprotective and cardioprotective activities. CAPE showed promising efficacy both in vitro and in vivo studies in animal models with minimum adverse effects. Its effectiveness was demonstrated in multiple target organs. Despite this fact, it has not been yet investigated as a protective agent or a potential therapy in humans. Investigation of CAPE efficacy in clinical trials is strongly encouraged to elucidate its therapeutic benefit for different human diseases after performing full preclinical toxicological studies and gaining more insights into its pharmacokinetics.
Collapse
Affiliation(s)
- Mai F Tolba
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | | | | | | | | |
Collapse
|
83
|
Ginhoux F, Lim S, Hoeffel G, Low D, Huber T. Origin and differentiation of microglia. Front Cell Neurosci 2013; 7:45. [PMID: 23616747 PMCID: PMC3627983 DOI: 10.3389/fncel.2013.00045] [Citation(s) in RCA: 607] [Impact Index Per Article: 50.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 03/30/2013] [Indexed: 12/30/2022] Open
Abstract
Microglia are the resident macrophage population of the central nervous system (CNS). Adequate microglial function is crucial for a healthy CNS. Microglia are not only the first immune sentinels of infection, contributing to both innate and adaptive immune responses locally, but are also involved in the maintenance of brain homeostasis. Emerging data are showing new and fundamental roles for microglia in the control of neuronal proliferation and differentiation, as well as in the formation of synaptic connections. While microglia have been studied for decades, a long history of experimental misinterpretation meant that their true origins remained debated. However, recent studies on microglial origin indicate that these cells in fact arise early during development from progenitors in the embryonic yolk sac (YS) that seed the brain rudiment and, remarkably, appear to persist there into adulthood. Here, we review the history of microglial cells and discuss the latest advances in our understanding of their origin, differentiation, and homeostasis, which provides new insights into their roles in health and disease.
Collapse
Affiliation(s)
- Florent Ginhoux
- Singapore Immunology Network, Agency for Science, Technology, and ResearchSingapore
| | - Shawn Lim
- Genome Institute Singapore, Agency for Science, Technology, and ResearchSingapore
| | - Guillaume Hoeffel
- Singapore Immunology Network, Agency for Science, Technology, and ResearchSingapore
| | - Donovan Low
- Singapore Immunology Network, Agency for Science, Technology, and ResearchSingapore
| | - Tara Huber
- Genome Institute Singapore, Agency for Science, Technology, and ResearchSingapore
- Department of Biological Science, National University of SingaporeSingapore
| |
Collapse
|
84
|
Busulfan conditioning enhances engraftment of hematopoietic donor-derived cells in the brain compared with irradiation. Mol Ther 2013; 21:868-76. [PMID: 23423338 DOI: 10.1038/mt.2013.29] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hematopoietic stem cell gene therapy for neurological disorders relies on transmigration of donor-derived monocytes to the brain, where they can engraft as microglia and deliver therapeutic proteins. Many mouse studies use whole-body irradiation to investigate brain transmigration pathways, but chemotherapy is generally used clinically. The current evidence for transmigration to the brain after chemotherapy is conflicting. We compared hematopoietic donor cell brain engraftment after bone marrow (BM) transplants in busulfan- or irradiation-conditioned mice. Significantly more donor-derived microglial cells engrafted posttransplant in busulfan-conditioned brain compared with the irradiated, in both the short and long term. Although total Iba-1(+) microglial content was increased in irradiated brain in the short term, it was similar between groups over long-term engraftment. MCP-1, a key regulator of monocyte transmigration, showed long-term elevation in busulfan-conditioned brain, whereas irradiated brains showed long-term elevation of the proinflammatory chemokine interleukin 1α (IL-1α), with increased in situ proliferation of resident microglia, and significant increases in the relative number of amoeboid activated microglia in the brain. This has implications for the choice of conditioning regimen to promote hematopoietic cell brain engraftment and the relevance of irradiation in mouse models of transplantation.
Collapse
|
85
|
Coquery N, Pannetier N, Farion R, Herbette A, Azurmendi L, Clarencon D, Bauge S, Josserand V, Rome C, Coll JL, Sun JS, Barbier EL, Dutreix M, Remy CC. Distribution and radiosensitizing effect of cholesterol-coupled Dbait molecule in rat model of glioblastoma. PLoS One 2012; 7:e40567. [PMID: 22815765 PMCID: PMC3398898 DOI: 10.1371/journal.pone.0040567] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 06/09/2012] [Indexed: 11/19/2022] Open
Abstract
Background Glioma is the most aggressive tumor of the brain and the most efficient treatments are based on radiotherapy. However, tumors are often resistant to radiotherapy due to an enhanced DNA repair activity. Short and stabilized DNA molecules (Dbait) have recently been proposed as an efficient strategy to inhibit DNA repair in tumor. Methodology/Principal Findings The distribution of three formulations of Dbait, (i) Dbait alone, (ii) Dbait associated with polyethylenimine, and (iii) Dbait linked with cholesterol (coDbait), was evaluated one day after intratumoral delivery in an RG2 rat glioma model. Dbait molecule distribution was assessed in the whole organ with 2D-FRI and in brain sections. CoDbait was chosen for further studies given its good retention in the brain, cellular localization, and efficacy in inducing the activation of DNA repair effectors. The radiosensitizing effect of coDbait was studied in four groups of rats bearing RG2-glioma: no treatment, radiotherapy only, coDbait alone, and CoDbait with radiotherapy. Treatment started 7 days after tumor inoculation and consisted of two series of treatment in two weeks: coDbait injection followed by a selective 6-Gy irradiation of the head. We evaluated the radiosensitizing effect using animal survival, tumor volume, cell proliferation, and vasculature characteristics with multiparametric MRI. CoDbait with radiotherapy improved the survival of rats bearing RG2-glioma by reducing tumor growth and cell proliferation without altering tumor vasculature. Conclusion/Significance coDbait is therefore a promising molecular therapy to sensitize glioma to radiotherapy.
Collapse
|
86
|
Shi HS, Gao X, Li D, Zhang QW, Wang YS, Zheng Y, Cai LL, Zhong RM, Rui A, Li ZY, Zheng H, Chen XC, Chen LJ. A systemic administration of liposomal curcumin inhibits radiation pneumonitis and sensitizes lung carcinoma to radiation. Int J Nanomedicine 2012; 7:2601-11. [PMID: 22679371 PMCID: PMC3368513 DOI: 10.2147/ijn.s31439] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Radiation pneumonitis (RP) is an important dose-limiting toxicity during thoracic radiotherapy. Previous investigations have shown that curcumin is used for the treatment of inflammatory conditions and cancer, suggesting that curcumin may prevent RP and sensitize cancer cells to irradiation. However, the clinical advancement of curcumin is limited by its poor water solubility and low bioavailability after oral administration. Here, a water-soluble liposomal curcumin system was developed to investigate its prevention and sensitizing effects by an intravenous administration manner in mice models. The results showed that liposomal curcumin inhibited nuclear factor-κB pathway and downregulated inflammatory factors including tumor necrosis factor-α, interleukin (IL)-6, IL-8, and transforming growth factor-β induced by thoracic irradiation. Furthermore, the combined treatment with liposomal curcumin and radiotherapy increased intratumoral apoptosis and microvessel responses to irradiation in vivo. The significantly enhanced inhibition of tumor growth also was observed in a murine lung carcinoma (LL/2) model. There were no obvious toxicities observed in mice. The current results indicate that liposomal curcumin can effectively mitigate RP, reduce the fibrosis of lung, and sensitize LL/2 cells to irradiation. This study also suggests that the systemic administration of liposomal curcumin is safe and deserves to be investigated for further clinical application.
Collapse
Affiliation(s)
- Hua-shan Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medicine School, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Viral-induced encephalitis initiates distinct and functional CD103+ CD11b+ brain dendritic cell populations within the olfactory bulb. Proc Natl Acad Sci U S A 2012; 109:6175-80. [PMID: 22474352 DOI: 10.1073/pnas.1203941109] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Dendritic cells (DC) are antigen-presenting cells found in both lymphoid and nonlymphoid organs, including the brain (bDC) of Cd11c/eyfp transgenic C57BL/6 mice. Using an intranasal vesicular stomatitis virus infection, we demonstrated that EYFP(+) cells amass in areas associated with viral antigens, take on an activated morphology, and project their processes into infected neuronal tissue within the olfactory bulb. These bDC separated into three EYFP(+) CD45(+) CD11b(+) populations, all but one being able to functionally promote both T lymphocyte proliferation and T(H)1 cytokine production. One population was shown to emanate from the brain and a second population was peripherally derived. The third population was of indeterminate origin, being both radiosensitive and not replenished by donor bone marrow. Finally, each EYFP(+) population contained CD11b(+) CD103(+) subpopulations and could be distinguished in terms of CD115, Gr-1, and Ly-6C expression, highlighting mucosal and monocyte-derived DC lineages.
Collapse
|
88
|
Kuffler DP. Hyperbaric oxygen therapy: can it prevent irradiation-induced necrosis? Exp Neurol 2012; 235:517-27. [PMID: 22465460 DOI: 10.1016/j.expneurol.2012.03.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 02/27/2012] [Accepted: 03/17/2012] [Indexed: 10/28/2022]
Abstract
Radiosurgery is an important non-invasive procedure for the treatment of tumors and vascular malformations. However, in addition to killing target tissues, cranial irradiation induces damage to adjacent healthy tissues leading to neurological deterioration in both pediatric and adult patients, which is poorly understood and insufficiently treatable. To minimize irradiation damage to healthy tissue, not the optimal therapeutic irradiation dose required to eliminate the target lesion is used but lower doses. Although the success rate of irradiation surgery is about 95%, 5% of patients suffer problems, most commonly neurological, that are thought to be a direct consequence of irradiation-induced inflammation. Although no direct correlation has been demonstrated, the appearance and disappearance of inflammation that develops following irradiation commonly parallel the appearance and disappearance of neurological side effects that are associated with the neurological function of the irradiated brain regions. These observations have led to the hypothesis that brain inflammation is causally related to the observed neurological side effects. Studies indicate that hyperbaric oxygen therapy (HBOT) applied after the appearance of irradiation-induced neurological side effects reduces the incidence and severity of those side effects. This may result from HBOT reducing inflammation, promoting angiogenesis, and influencing other cellular functions thereby suppressing events that cause the neurological side effects. However, it would be significantly better for the patient if rather than waiting for neurological side effects to become manifest they could be avoided. This review examines irradiation-induced neurological side effects, methods that minimize or resolve those side effects, and concludes with a discussion of whether HBOT applied following irradiation, but before manifestation of neurological side effects may prevent or reduce the appearance of irradiation-induced neurological side effects.
Collapse
Affiliation(s)
- Damien P Kuffler
- Institute of Neurobiology, University of Puerto Rico, Medical Sciences Campus, Puerto Rico.
| |
Collapse
|
89
|
Akyol S, Ginis Z, Armutcu F, Ozturk G, Yigitoglu MR, Akyol O. The potential usage of caffeic acid phenethyl ester (CAPE) against chemotherapy-induced and radiotherapy-induced toxicity. Cell Biochem Funct 2012; 30:438-43. [PMID: 22431158 DOI: 10.1002/cbf.2817] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2011] [Revised: 01/26/2012] [Accepted: 02/06/2012] [Indexed: 11/09/2022]
Abstract
Protection of the patients against the side effects of chemotherapy and radiotherapy regimens has attracted increasing interest of clinicians and practitioners. Caffeic acid phenethyl ester (CAPE), which is extracted from the propolis of honeybee hives as an active component, specifically inhibits nuclear factor κB at micromolar concentrations and show ability to stop 5-lipoxygenase-catalysed oxygenation of linoleic acid and arachidonic acid. CAPE has antiinflammatory, antiproliferative, antioxidant, cytostatic, antiviral, antibacterial, antifungal and antineoplastic properties. The purpose of this review is to summarize in vivo and in vitro usage of CAPE to prevent the chemotherapy-induced and radiotherapy-induced damages and side effects in experimental animals and to develop a new approach for the potential usage of CAPE in clinical trial as a protective agent during chemotherapy and radiotherapy regimens.
Collapse
Affiliation(s)
- Sumeyya Akyol
- Department of Biochemistry, Fatih University Medical School, Ankara, Turkey
| | | | | | | | | | | |
Collapse
|
90
|
Gallet P, Phulpin B, Merlin JL, Leroux A, Bravetti P, Mecellem H, Tran N, Dolivet G. Long-term alterations of cytokines and growth factors expression in irradiated tissues and relation with histological severity scoring. PLoS One 2011; 6:e29399. [PMID: 22216271 PMCID: PMC3245280 DOI: 10.1371/journal.pone.0029399] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 11/28/2011] [Indexed: 02/06/2023] Open
Abstract
Purpose Beside its efficacy in cancer treatment, radiotherapy induces degeneration of healthy tissues within the irradiated area. The aim of this study was to analyze the variations of proinflammatory (IL-1α, IL-2, IL-6, TNF-α, IFN-γ), profibrotic (TGF-β1), proangiogneic (VEGF) and stem cell mobilizing (GM-CSF) cytokines and growth factors in an animal model of radiation-induced tissue degeneration. Materials and Methods 24 rats were irradiated unilaterally on the hindlimb at a monodose of 30 Gy. Six weeks (n = 8), 6 months (n = 8) and 1 year (n = 8) after irradiation the mediators expression in skin and muscle were analyzed using Western blot and the Bio-Plex® protein array (BPA) technology. Additional histological severity for fibrosis, inflammation, vascularity and cellularity alterations scoring was defined from histology and immnunohistochemistry analyses. Results A significant increase of histological severity scoring was found in irradiated tissue. Skin tissues were more radio-sensitive than muscle. A high level of TGF-β1 expression was found throughout the study and a significant relation was evidenced between TGF-β1 expression and fibrosis scoring. Irradiated tissue showed a chronic inflammation (IL-2 and TNF-α significantly increased). Moreover a persistent expression of GM-CSF and VEGF was found in all irradiated tissues. The vascular score was related to TGF-β1 expression and the cellular alterations score was significantly related with the level of IL-2, VEGF and GM-CSF. Conclusion The results achieved in the present study underline the complexity and multiplicity of radio-induced alterations of cytokine network. It offers many perspectives of development, for the comprehension of the mechanisms of late injuries or for the histological and molecular evaluation of the mode of action and the efficacy of rehabilitation techniques.
Collapse
Affiliation(s)
- Patrice Gallet
- EA4421 SiGReTO Nancy University, Faculty of Medicine, Vandoeuvre-lès-Nancy, France
| | - Bérengère Phulpin
- EA4421 SiGReTO Nancy University, Faculty of Medicine, Vandoeuvre-lès-Nancy, France
- Head and Neck Surgery and Dental Units, Oncologic Surgery Department, Centre Alexis Vautrin, Vandoeuvre-lès-Nancy, France
- * E-mail:
| | - Jean-Louis Merlin
- EA4421 SiGReTO Nancy University, Faculty of Medicine, Vandoeuvre-lès-Nancy, France
- Pathology and Tumor Biology Department, Centre Alexis Vautrin, Vandoeuvre-lès- Nancy, France
| | - Agnès Leroux
- EA4421 SiGReTO Nancy University, Faculty of Medicine, Vandoeuvre-lès-Nancy, France
- Pathology and Tumor Biology Department, Centre Alexis Vautrin, Vandoeuvre-lès- Nancy, France
| | - Pierre Bravetti
- Oral surgery department, Faculty of Dentistry, Nancy University, Nancy, France
| | - Hinda Mecellem
- Radiotherapy Department, Centre Alexis Vautrin, Vandoeuvre-lès-Nancy, France
| | - Nguyen Tran
- School of Surgery, INSERM U961, Faculty of Medicine, Nancy University, Vandoeuvre-lès-Nancy, France
- INSERM U961, Faculty of Medicine, Nancy University, Vandoeuvre-lès-Nancy, France
| | - Gilles Dolivet
- EA4421 SiGReTO Nancy University, Faculty of Medicine, Vandoeuvre-lès-Nancy, France
- Head and Neck Surgery and Dental Units, Oncologic Surgery Department, Centre Alexis Vautrin, Vandoeuvre-lès-Nancy, France
| |
Collapse
|
91
|
Dicker AP, Kari G, Rodeck U. Radiation-Induced Toxicity and Radiation Response Modifiers in Zebrafish. Zebrafish 2011. [DOI: 10.1002/9781118102138.ch21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
92
|
Distinct and non-redundant roles of microglia and myeloid subsets in mouse models of Alzheimer's disease. J Neurosci 2011; 31:11159-71. [PMID: 21813677 DOI: 10.1523/jneurosci.6209-10.2011] [Citation(s) in RCA: 275] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Mononuclear phagocytes are important modulators of Alzheimer's disease (AD), but the specific functions of resident microglia, bone marrow-derived mononuclear cells, and perivascular macrophages have not been resolved. To elucidate the spatiotemporal roles of mononuclear phagocytes during disease, we targeted myeloid cell subsets from different compartments and examined disease pathogenesis in three different mouse models of AD (APP(swe/PS1), APP(swe), and APP23 mice). We identified chemokine receptor 2 (CCR2)-expressing myeloid cells as the population that was preferentially recruited to β-amyloid (Aβ) deposits. Unexpectedly, AD brains with dysfunctional microglia and devoid of parenchymal bone marrow-derived phagocytes did not show overt changes in plaque pathology and Aβ load. In contrast, restriction of CCR2 deficiency to perivascular myeloid cells drastically impaired β-amyloid clearance and amplified vascular Aβ deposition, while parenchymal plaque deposition remained unaffected. Together, our data advocate selective functions of CCR2-expressing myeloid subsets, which could be targeted specifically to modify disease burden in AD.
Collapse
|
93
|
Soucy KG, Lim HK, Kim JH, Oh Y, Attarzadeh DO, Sevinc B, Kuo MM, Shoukas AA, Vazquez ME, Berkowitz DE. HZE ⁵⁶Fe-ion irradiation induces endothelial dysfunction in rat aorta: role of xanthine oxidase. Radiat Res 2011; 176:474-85. [PMID: 21787183 DOI: 10.1667/rr2598.1] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Ionizing radiation has been implicated in the development of significant cardiovascular complications. Since radiation exposure is associated with space exploration, astronauts are potentially at increased risk of accelerated cardiovascular disease. This study investigated the effect of high atomic number, high-energy (HZE) iron-ion radiation on vascular and endothelial function as a model of space radiation. Rats were exposed to a single whole-body dose of iron-ion radiation at doses of 0, 0.5 or 1 Gy. In vivo aortic stiffness and ex vivo aortic tension responses were measured 6 and 8 months after exposure as indicators of chronic vascular injury. Rats exposed to 1 Gy iron ions demonstrated significantly increased aortic stiffness, as measured by pulse wave velocity. Aortic rings from irradiated rats exhibited impaired endothelial-dependent relaxation consistent with endothelial dysfunction. Acute xanthine oxidase (XO) inhibition or reactive oxygen species (ROS) scavenging restored endothelial-dependent responses to normal. In addition, XO activity was significantly elevated in rat aorta 4 months after whole-body irradiation. Furthermore, XO inhibition, initiated immediately after radiation exposure and continued until euthanasia, completely inhibited radiation-dependent XO activation. ROS production was elevated after 1 Gy irradiation while production of nitric oxide (NO) was significantly impaired. XO inhibition restored NO and ROS production. Finally, dietary XO inhibition preserved normal endothelial function and vascular stiffness after radiation exposure. These results demonstrate that radiation induced XO-dependent ROS production and nitroso-redox imbalance, leading to chronic vascular dysfunction. As a result, XO is a potential target for radioprotection. Enhancing the understanding of vascular radiation injury could lead to the development of effective methods to ameliorate radiation-induced vascular damage.
Collapse
Affiliation(s)
- Kevin G Soucy
- Biomedical Engineering, Johns Hopkins Medical Institutions, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Kim W, Seong KM, Youn B. Phenylpropanoids in radioregulation: double edged sword. Exp Mol Med 2011; 43:323-33. [PMID: 21483230 PMCID: PMC3128910 DOI: 10.3858/emm.2011.43.6.034] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2011] [Indexed: 11/04/2022] Open
Abstract
Radiotherapy, frequently used for treatment of solid tumors, carries two main obstacles including acquired radioresistance in cancer cells during radiotherapy and normal tissue injury. Phenylpropanoids, which are naturally occurring phytochemicals found in plants, have been identified as potential radiotherapeutic agents due to their anti-cancer activity and relatively safe levels of cytotoxicity. Various studies have proposed that these compounds could not only sensitize cancer cells to radiation resulting in inhibition of growth and cell death but also protect normal cells against radiation-induced damage. This review is intended to provide an overview of recent investigations on the usage of phenylpropanoids in combination with radiotherapy in cancer treatment.
Collapse
Affiliation(s)
- Wanyeon Kim
- College of Natural Sciences Department of Biological Sciences Pusan National University Busan 609-735, Korea
| | | | | |
Collapse
|
95
|
Biton S, Ashkenazi A. NEMO and RIP1 control cell fate in response to extensive DNA damage via TNF-α feedforward signaling. Cell 2011; 145:92-103. [PMID: 21458669 DOI: 10.1016/j.cell.2011.02.023] [Citation(s) in RCA: 312] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Revised: 01/21/2011] [Accepted: 02/04/2011] [Indexed: 11/30/2022]
Abstract
Upon DNA damage, ataxia telangiectasia mutated (ATM) kinase triggers multiple events to promote cell survival and facilitate repair. If damage is excessive, ATM stimulates cytokine secretion to alert neighboring cells and apoptosis to eliminate the afflicted cell. ATM augments cell survival by activating nuclear factor (NF)-κB; however, how ATM induces cytokine production and apoptosis remains elusive. Here we uncover a p53-independent mechanism that transmits ATM-driven cytokine and caspase signals upon strong genotoxic damage. Extensive DNA lesions stimulated two sequential NF-κB activation phases, requiring ATM and NEMO/IKK-γ: The first phase induced TNF-α-TNFR1 feedforward signaling, promoting the second phase and driving RIP1 phosphorylation. In turn, RIP1 kinase triggered JNK3/MAPK10-dependent interleukin-8 secretion and FADD-mediated proapoptotic caspase-8 activation. Thus, in the context of excessive DNA damage, ATM employs NEMO and RIP1 kinase through autocrine TNF-α signaling to switch on cytokine production and caspase activation. These results shed light on cell-fate regulation by ATM.
Collapse
Affiliation(s)
- Sharon Biton
- Department of Molecular Oncology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | | |
Collapse
|
96
|
Oh JH, Craft JM, Townsend R, Deasy JO, Bradley JD, El Naqa I. A bioinformatics approach for biomarker identification in radiation-induced lung inflammation from limited proteomics data. J Proteome Res 2011; 10:1406-15. [PMID: 21226504 PMCID: PMC3127583 DOI: 10.1021/pr101226q] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Many efforts have been made to discover novel bio-markers for early disease detection in oncology. However, the lack of efficient computational strategies impedes the discovery of disease-specific biomarkers for better understanding and management of treatment outcomes. In this study, we propose a novel graph-based scoring function to rank and identify the most robust biomarkers from limited proteomics data. The proposed method measures the proximity between candidate proteins identified by mass spectrometry (MS) analysis utilizing prior reported knowledge in the literature. Recent advances in mass spectrometry provide new opportunities to identify unique biomarkers from peripheral blood samples in complex treatment modalities such as radiation therapy (radiotherapy), which enables early disease detection, disease progression monitoring, and targeted intervention. Specifically, the dose-limiting role of radiation-induced lung injury known as radiation pneumonitis (RP) in lung cancer patients receiving radiotherapy motivates the search for robust predictive biomarkers. In this case study, plasma from 26 locally advanced non-small cell lung cancer (NSCLC) patients treated with radiotherapy in a longitudinal 3 × 3 matched-control cohort was fractionated using in-line, sequential multiaffinity chromatography. The complex peptide mixtures from endoprotease digestions were analyzed using comparative, high-resolution liquid chromatography (LC)-MS to identify and quantify differential peptide signals. Through analysis of survey mass spectra and annotations of peptides from the tandem spectra, we found candidate proteins that appear to be associated with RP. On the basis of the proposed methodology, α-2-macroglobulin (α2M) was unambiguously ranked as the top candidate protein. As independent validation of this candidate protein, enzyme-linked immunosorbent assay (ELISA) experiments were performed on independent cohort of 20 patients' samples resulting in early significant discrimination between RP and non-RP patients (p = 0.002). These results suggest that the proposed methodology based on longitudinal proteomics analysis and a novel bioinformatics ranking algorithm is a potentially promising approach for the challenging problem of identifying relevant biomarkers in sample-limited clinical applications.
Collapse
Affiliation(s)
| | | | | | | | | | - Issam El Naqa
- To whom correspondence should be addressed Phone: +1 (314)362 0129. Fax: +1 (314)362 8521
| |
Collapse
|
97
|
Wolff HA, Rolke D, Rave-Fränk M, Schirmer M, Eicheler W, Doerfler A, Hille A, Hess CF, Matthias C, Rödel RMW, Christiansen H. Analysis of chemokine and chemokine receptor expression in squamous cell carcinoma of the head and neck (SCCHN) cell lines. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2011; 50:145-154. [PMID: 21085979 PMCID: PMC3040826 DOI: 10.1007/s00411-010-0341-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Accepted: 11/01/2010] [Indexed: 05/30/2023]
Abstract
The purpose of this work was to analyze chemokine and chemokine receptor expression in untreated and in irradiated squamous cell carcinoma of the head and neck (SCCHN) tumor cell lines, aiming at the establishment of assays to test for the relevance of chemokine and chemokine receptor expression in the response of SCCHN to radiotherapy and radiochemotherapy. Five low passage and 10 established SCCHN lines, as well as two normal cell lines, were irradiated at 2 Gy or sham-irradiated, and harvested between 1 and 48 h after treatment. For chemokines with CC and CXC structural motifs and their receptors, transcript levels of target and reference genes were quantified relatively by real-time PCR. In addition, CXCL1 and CXCL12 protein expression was analyzed by ELISA. A substantial variation in chemokine and chemokine receptor expression between SCCHN was detected. Practically, all cell lines expressed CCL5 and CCL20, while CCL2 was expressed in normal cells and in some of the tumor cell lines. CXCL1, CXCL2, CXCL3, CXCL10, and CXCL11 were expressed in the vast majority of the cell lines, while the expression of CXCL9 and CXCL12 was restricted to fibroblasts and few tumor cell lines. None of the analyzed cell lines expressed the chemokines CCL3, CCL4, or CCL19. Of the receptors, transcript expression of CCR1, CCR2, CCR3, CCR5, CCR7, CCXR2, and CCXR3 was not detected, and CCR6, CXCR1, and CXCR4 expression was restricted to few tumor cells. Radiation caused up- and down-regulation with respect to chemokine expressions, while for chemokine receptor expressions down-regulations were prevailing. CXCL1 and CXCL12 protein expression corresponded well with the mRNA expression. We conclude that the substantial variation in chemokine and chemokine receptor expression between SCCHN offer opportunities for the establishment of assays to test for the relevance of chemokine and chemokine receptor expression in the response of SCCHN to radiotherapy and radiochemotherapy.
Collapse
MESH Headings
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/radiotherapy
- Cell Line, Tumor
- Chemokines/genetics
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/radiation effects
- Head and Neck Neoplasms/drug therapy
- Head and Neck Neoplasms/genetics
- Head and Neck Neoplasms/pathology
- Head and Neck Neoplasms/radiotherapy
- Humans
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Chemokine/genetics
- Reproducibility of Results
Collapse
Affiliation(s)
- Hendrik A. Wolff
- Department of Radiotherapy and Radiation Oncology, Universitätsmedizin Göttingen, Göttingen, Germany
| | - David Rolke
- Department of Radiotherapy and Radiation Oncology, Universitätsmedizin Göttingen, Göttingen, Germany
| | - Margret Rave-Fränk
- Department of Radiotherapy and Radiation Oncology, Universitätsmedizin Göttingen, Göttingen, Germany
| | - Markus Schirmer
- Department of Pharmacology, Universitätsmedizin Göttingen, Göttingen, Germany
| | - Wolfgang Eicheler
- Department of Radiation Oncology, OncoRay-Center for Radiation Research in Oncology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Annegret Doerfler
- Department of Radiation Oncology, OncoRay-Center for Radiation Research in Oncology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Andrea Hille
- Department of Radiotherapy and Radiation Oncology, Universitätsmedizin Göttingen, Göttingen, Germany
| | - Clemens F. Hess
- Department of Radiotherapy and Radiation Oncology, Universitätsmedizin Göttingen, Göttingen, Germany
| | - Christoph Matthias
- Department of Otorhinolaryngology-Head and Neck Surgery, Universitätsmedizin Göttingen, Göttingen, Germany
| | - Ralph M. W. Rödel
- Department of Otorhinolaryngology-Head and Neck Surgery, Universitätsmedizin Göttingen, Göttingen, Germany
| | - Hans Christiansen
- Department of Radiotherapy and Radiation Oncology, Universitätsmedizin Göttingen, Göttingen, Germany
| |
Collapse
|
98
|
El-ghazaly MA, Abd el-naby DH, Khayyal MT. The influence of irradiation on the potential chondroprotective effect of aqueous extract of propolis in rats. Int J Radiat Biol 2010; 87:254-62. [DOI: 10.3109/09553002.2011.530337] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
99
|
Chen MF, Chen WC, Lai CH, Hung CH, Liu KC, Cheng YH. Predictive factors of radiation-induced skin toxicity in breast cancer patients. BMC Cancer 2010; 10:508. [PMID: 20860847 PMCID: PMC2955039 DOI: 10.1186/1471-2407-10-508] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Accepted: 09/23/2010] [Indexed: 11/21/2022] Open
Abstract
Background To assess the factors affecting the incidence of radiation-induced dermatitis in breast cancer patients treated with adjuvant 3 D conformal radiotherapy by the analysis of dosimetry and topical treatments. Methods Between September 2002 and July 2009, 158 breast cancer patients were treated with adjuvant 3 D conformal radiotherapy after undergoing surgery. Before November 2006, 90 patients were subjected to therapeutic skin care group and topical corticosteroid therapy was used for acute radiation dermatitis. Thereafter, 68 patients received prophylactic topical therapy from the beginning of radiotherapy. The two groups did not differ significantly in respect of clinical and treatment factors. Furthermore, the possible mechanisms responsible for the effects of topical treatment on radiation-induced dermatitis were investigated in vivo. Results The incidence of radiation-induced moist desquamation was 23% across 158 patients. Higher volume receiving 107% of prescribed dose within PTV (PTV-V107%; >28.6%) and volume receiving 110% of prescribed dose within treated volume (TV-V110%; > 5.13%), and no prophylactic topical therapy for irradiated skin, were associated with higher incidence of acute radiation dermatitis. The protective effect of prophylactic topical treatment was more pronounced in patients with TV-V110% > 5.13%. Furthermore, using irradiated mice, we demonstrated that topical steroid cream significantly attenuated irradiation-induced inflammation, causing a decrease in expression of inflammatory cytokines and TGF-beta 1. Conclusion TV-V110% > 5.13% may be an important predictor for radiation induced dermatitis. Prophylactic topical treatment for irradiated skin can significantly improve the tolerance of skin to adjuvant radiotherapy, especially for patients with higher TV-V110%.
Collapse
Affiliation(s)
- Miao-Fen Chen
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Chia-Yi, Taiwan.
| | | | | | | | | | | |
Collapse
|
100
|
Abd-El-Fattah AA, El-Sawalhi MM, Rashed ER, El-Ghazaly MA. Possible role of vitamin E, coenzyme Q10 and rutin in protection against cerebral ischemia/reperfusion injury in irradiated rats. Int J Radiat Biol 2010; 86:1070-8. [PMID: 20712430 DOI: 10.3109/09553002.2010.501844] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE To investigate the possible role of vitamin E, coenzyme Q10 and rutin in ameliorating the biochemical changes in brain and serum induced by cerebral ischemia/reperfusion (I/R) in whole body γ-irradiated rats. MATERIALS AND METHODS Cerebral ischemia was induced in male Wistar rats (either irradiated or non-irradiated) followed by reperfusion. RESULTS I/R increased brain content of malondialdehyde (MDA) and depleted its glutathione (GSH) content with a compensatory elevation in cytosolic activities of glutathione peroxidase (GPx) and glutathione reductase (GR) enzymes. It also raised brain cytosolic lactate dehydrogenase (LDH) activity and calcium (Ca(2+)) level. Furthermore, I/R provoked an inflammatory response reflected by an increment in serum levels of the proinflammatory cytokines tumour necrosis factor-α (TNF-α) and interlukin-1β (IL-1β). Moreover, induction of I/R in irradiated rats resulted in a further increase in brain oxidative stress and cytosolic LDH activity, disturbed brain Ca(2+) homeostasis and exaggerated the inflammatory reaction. During irradiation, administration of each of vitamin E, coenzyme Q10 (CoQ10) and rutin to irradiated rats before induction of I/R, alleviated the brain oxidative stress. Moreover, these antioxidants caused attenuation of the rise of the cytosolic activities of GPx and GR. A lowering effect of the cytosolic LDH activity and Ca(2+) level were caused by treatment with antioxidants. Each of vitamin E and rutin revealed an anti-inflammatory action of these antioxidants, while CoQ10 had no effect on serum levels of TNF-α and IL-1β. CONCLUSION These findings indicate that supplementation with either vitamin E, CoQ10 or rutin ameliorated most of the biochemical changes induced by I/R in irradiated rat brain and serum.
Collapse
Affiliation(s)
- Amal A Abd-El-Fattah
- Drug Radiation Research Department, National Centre for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | | | | | | |
Collapse
|