1
|
Liu D, Wei M, Fang Y, Yuan T, Sun Y, Xie H, Yan W, Yuan B, Zhuang B, Jin Y. Oral colon-retentive inulin gels protect against radiation-induced hematopoietic and gastrointestinal injury by improving gut homeostasis. Int J Biol Macromol 2024; 292:139199. [PMID: 39730057 DOI: 10.1016/j.ijbiomac.2024.139199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024]
Abstract
Ionizing radiation-induced injury often occurs in nuclear accidents or large-dose radiotherapy, leading to acute radiation syndromes characterized by hematopoietic and gastrointestinal injuries even to death. However, current radioprotective drugs are only used in hospitals with unavoidable side effects. Here, we heated the aqueous solution of inulin, a polysaccharide dietary fiber, forming colon-retentive gel as a radiation protector in radiotherapy. Mouse models were established after 60Co γ-ray irradiation of the total body or abdomen. Inulin gels were orally administered to the mice every day from 3 days pre-radiation to 3 days post-radiation. The hematopoietic system was well protected with good blood cell recovery and cell proliferation in the femur and spleen. Oral inulin gels increased the relative abundances of key commensal microorganisms including f_Lachnospiraceae, Akkermansia, Blautia, and short-chain fatty acid metabolites. The secretion of the anti-inflammation cytokines IL-22 and IL-10 in the intestinal cells also increased. Similarly, the expression of the tight junction proteins claudin-1 and occludin in the gut mucosa was affected. In an orthotopic murine colorectal cancer model, oral inulin gels followed by 10-Gy abdomen radiation improved the radiotherapy efficiency with low attenuated radiation injury. Taken the data together, these results suggest that oral inulin gels are a bioactive material against ionizing radiation-induced injury.
Collapse
Affiliation(s)
- Dongdong Liu
- Beijing Institute of Radiation Medicine, Beijing 100850, China; China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing 100068, China
| | - Meng Wei
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yubao Fang
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Tianyu Yuan
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yingbao Sun
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Hua Xie
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Wenrui Yan
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Bochuan Yuan
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Bo Zhuang
- Institute of NBC Defense, Beijing 102205, China.
| | - Yiguang Jin
- Beijing Institute of Radiation Medicine, Beijing 100850, China.
| |
Collapse
|
2
|
Wang L, Lin B, Zhai M, Hull L, Cui W, Xiao M. Endothelial Dysfunction and Impaired Wound Healing Following Radiation Combined Skin Wound Injury. Int J Mol Sci 2024; 25:12498. [PMID: 39684207 DOI: 10.3390/ijms252312498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Currently, there are no U.S. Food and Drug Administration (FDA)-approved medical countermeasures (MCMs) for radiation combined injury (RCI), partially due to limited understanding of its mechanisms. Our previous research suggests that endothelial dysfunction may contribute to a poor prognosis of RCI. In this study, we demonstrated an increased risk of mortality, body weight loss, and delayed skin wound healing in RCI mice compared to mice with skin wounds alone or radiation injury (RI) 30 days post-insult. Furthermore, we evaluated biomarkers of endothelial dysfunction, inflammation, and impaired wound healing in mice at early time points after RCI. Mice were exposed to 9.0 Gy total-body irradiation (TBI) followed by skin wound. Samples were collected on days 3, 7, and 14 post-TBI. Endothelial dysfunction markers were measured by ELISA, and skin wound healing was assessed histologically. Our results show that endothelial damage and inflammation are more severe and persistent in the RCI compared to the wound-alone group. Additionally, RCI impairs granulation tissue formation, reduces myofibroblast presence, and delays collagen deposition, correlating with more severe endothelial damage. TGF signaling may play a key role in this impaired healing. These findings suggest that targeting the endothelial dysfunction and TGF-β pathways may provide potential therapeutic strategies for improving delayed wound healing in RCI, which could subsequently influence outcomes such as survival after RCI.
Collapse
Affiliation(s)
- Li Wang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Henry M Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Bin Lin
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Henry M Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Min Zhai
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Henry M Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Lisa Hull
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Henry M Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Wanchang Cui
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Henry M Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Mang Xiao
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| |
Collapse
|
3
|
Luan J, Feng X, Du Y, Yang D, Geng C. Medium-chain fatty acid triglycerides improve feed intake and oxidative stress of finishing bulls by regulating ghrelin concentration and gastrointestinal tract microorganisms and rumen metabolites. MICROBIOME 2024; 12:230. [PMID: 39511583 PMCID: PMC11542207 DOI: 10.1186/s40168-024-01946-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 10/09/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND As a feed additive, medium-chain fatty acids (MCFAs)/medium-chain fatty acid triglycerides (MCTs) have been used in ruminant production, but mostly added in the form of mixed esters. Studies have shown that MCTs may have a positive effect on feed intake or oxidative stress in animals, but it is unclear which MCT could play a role, and the mechanism has not been elucidated. In this study, the effects of individual MCT on growth performance, serum intake-related hormones, and oxidative stress indices in finishing bulls were investigated and further studied the effects of MCT supplementation on gastrointestinal tract bacteria and rumen fluid metabolomics. RESULTS Four ruminally fistulated Yanbian cattle (bulls) were selected in 4 × 4 Latin square designs and allocated to four treatment groups: a control group (CON) fed a basal diet (total mixed ration, TMR), three groups fed a basal diet supplemented with 60 g/bull/day glycerol monocaprylin (GMC, C8), glycerol monodecanoate (GMD, C10), and glycerol monolaurate (GML, C12), respectively. Compared with the CON group, GMD tended to increase the dry matter intake (DMI) of finishing bulls (P = 0.069). Compared with the CON group, GMD significantly increased the concentration of ghrelin O-acyl transferase (GOAT), total ghrelin (TG), acylated ghrelin (AG), and orexins (P < 0.05) and significantly decreased the concentrations of hydrogen peroxide (H2O2), malondialdehyde, reactive oxygen species (ROS), and lipopolysaccharides (LPS) in the serum of finishing bulls (P < 0.05). Compared with the CON group, GMD and GML significantly increased the concentrations of total antioxidant capacity (T-AOC), catalase, glutathione peroxidase (GSH-PX), glutathione reductase (GR), and nitric oxide (NO) in the serum of finishing bulls (P < 0.05). Compared with the CON group, there were 5, 14, and 6 significantly different bacteria in the rumen digesta in the C8, C10, and C12 groups, respectively; there were 3, 10, and 5 significantly different bacteria in the rumen fluid in the C8, C10, and C12 groups, respectively; and only one differential bacteria (genus level) in the feces among the four treatment groups. Compared with the CON group, there were 3, 14, and 15 significantly differential metabolites identified under positive ionization mode in the C8, C10, and C12 groups, respectively, while under negative ionization mode were 3, 11 and 14, respectively. Correlation analysis showed that there was a significant correlation between DMI, GOAT, AG, GSH-PX, LPS, gastrointestinal tract bacteria, and rumen fluid metabolites. CONCLUSIONS Our findings revealed that different types of MCTs have different application effects in ruminants. Among them, GMD may improve the feed intake of finishing bulls by stimulating the secretion of AG. GMD and GML may change gastrointestinal tract microorganisms and produce specific rumen metabolites to improve the oxidative stress of finishing bulls, and ghrelin may also be involved. This study enlightens the potential mechanisms by which MCT improves feed intake and oxidative stress in finishing bulls. Video Abstract.
Collapse
Affiliation(s)
- Jiaming Luan
- Agricultural College, Yanbian University, Yanji, 133002, China
| | - Xin Feng
- Agricultural College, Yanbian University, Yanji, 133002, China
| | - Yunlong Du
- Agricultural College, Yanbian University, Yanji, 133002, China
| | - Dongxu Yang
- Agricultural College, Yanbian University, Yanji, 133002, China
| | - Chunyin Geng
- Agricultural College, Yanbian University, Yanji, 133002, China.
- Engineering Research Center of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Yanbian University, Yanji, 133002, China.
| |
Collapse
|
4
|
Liao SS, Zhang LL, Zhang YG, Luo J, Kadier T, Ding K, Chen R, Meng QT. Ghrelin alleviates intestinal ischemia-reperfusion injury by activating the GHSR-1α/Sirt1/FOXO1 pathway. FASEB J 2024; 38:e23681. [PMID: 38814725 DOI: 10.1096/fj.202302155rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 04/15/2024] [Accepted: 05/07/2024] [Indexed: 06/01/2024]
Abstract
Ischemia-reperfusion (IR) injury is primarily characterized by the restoration of blood flow perfusion and oxygen supply to ischemic tissue and organs, but it paradoxically leads to tissue injury aggravation. IR injury is a challenging pathophysiological process that is difficult to avoid clinically and frequently occurs during organ transplantation, surgery, shock resuscitation, and other processes. The major causes of IR injury include increased levels of free radicals, calcium overload, oxidative stress, and excessive inflammatory response. Ghrelin is a newly discovered brain-intestinal peptide with anti-inflammatory and antiapoptotic effects that improve blood supply. The role and mechanism of ghrelin in intestinal ischemia-reperfusion (IIR) injury remain unclear. We hypothesized that ghrelin could attenuate IIR-induced oxidative stress and apoptosis. To investigate this, we established IIR by using a non-invasive arterial clip to clamp the root of the superior mesenteric artery (SMA) in mice. Ghrelin was injected intraperitoneally at a dose of 50 μg/kg 20 min before IIR surgery, and [D-Lys3]-GHRP-6 was injected intraperitoneally at a dose of 12 nmol/kg 20 min before ghrelin injection. We mimicked the IIR process with hypoxia-reoxygenation (HR) in Caco-2 cells, which are similar to intestinal epithelial cells in structure and biochemistry. Our results showed that ghrelin inhibited IIR/HR-induced oxidative stress and apoptosis by activating GHSR-1α. Moreover, it was found that ghrelin activated the GHSR-1α/Sirt1/FOXO1 signaling pathway. We further inhibited Sirt1 and found that Sirt1 was critical for ghrelin-mediated mitigation of IIR/HR injury. Overall, our data suggest that pretreatment with ghrelin reduces oxidative stress and apoptosis to attenuate IIR/HR injury by binding with GHSR-1α to further activate Sirt1.
Collapse
Affiliation(s)
- Shi-Shi Liao
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Le-le Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi-Guo Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jie Luo
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tulanisa Kadier
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ke Ding
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rong Chen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Anesthesiology, East Hospital, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qing-Tao Meng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Anesthesiology, East Hospital, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
5
|
Horseman TS, Frank AM, Cannon G, Zhai M, Olson MG, Lin B, Li X, Hull L, Xiao M, Kiang JG, Burmeister DM. Effects of combined ciprofloxacin and Neulasta therapy on intestinal pathology and gut microbiota after high-dose irradiation in mice. Front Public Health 2024; 12:1365161. [PMID: 38807988 PMCID: PMC11130442 DOI: 10.3389/fpubh.2024.1365161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 03/20/2024] [Indexed: 05/30/2024] Open
Abstract
Introduction Treatments that currently exist in the strategic national stockpile for acute radiation syndrome (ARS) focus on the hematopoietic subsyndrome, with no treatments on gastrointestinal (GI)-ARS. While the gut microbiota helps maintain host homeostasis by mediating GI epithelial and mucosal integrity, radiation exposure can alter gut commensal microbiota which may leave the host susceptible to opportunistic pathogens and serious sequelae such as sepsis. To mitigate the effects of hematopoietic ARS irradiation, currently approved treatments exist in the form of colony stimulating factors and antibiotics: however, there are few studies examining how these therapeutics affect GI-ARS and the gut microbiota. The aim of our study was to examine the longitudinal effects of Neulasta and/or ciprofloxacin treatment on the gut microbiota after exposure to 9.5 Gy 60Co gamma-radiation in mice. Methods The gut microbiota of vehicle and drug-treated mice exposed to sham or gamma-radiation was characterized by shotgun sequencing with alpha diversity, beta diversity, and taxonomy analyzed on days 2, 4, 9, and 15 post-irradiation. Results No significant alpha diversity differences were observed following radiation, while beta diversity shifts and taxonomic profiles revealed significant alterations in Akkermansia, Bacteroides, and Lactobacillus. Ciprofloxacin generally led to lower Shannon diversity and Bacteroides prevalence with increases in Akkermansia and Lactobacillus compared to vehicle treated and irradiated mice. While Neulasta increased Shannon diversity and by day 9 had more similar taxonomic profiles to sham than ciprofloxacin-or vehicle-treated irradiated animals. Combined therapy of Neulasta and ciprofloxacin induced a decrease in Shannon diversity and resulted in unique taxonomic profiles early post-irradiation, returning closer to vehicle-treated levels over time, but persistent increases in Akkermansia and Bacteroides compared to Neulasta alone. Discussion This study provides a framework for the identification of microbial elements that may influence radiosensitivity, biodosimetry and the efficacy of potential therapeutics. Moreover, increased survival from H-ARS using these therapeutics may affect the symptoms and appearance of what may have been subclinical GI-ARS.
Collapse
Affiliation(s)
- Timothy S. Horseman
- School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Andrew M. Frank
- School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Georgetta Cannon
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Min Zhai
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Matthew G. Olson
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Bin Lin
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Xianghong Li
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Lisa Hull
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Mang Xiao
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Juliann G. Kiang
- School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - David M. Burmeister
- School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
6
|
Bolduc DL, Cary LH, Kiang JG, Kurada L, Kumar VP, Edma SA, Olson MG, Vergara VB, Bistline DD, Reese M, Kenchegowda D, Hood M, Korotcov A, Jaiswal S, Blakely WF. Natural-history Characterization of a Murine Partial-body Irradiation Model System: Establishment of a Multiple-Parameter Based GI-ARS Severity-Scoring System. Radiat Res 2024; 201:406-417. [PMID: 38319684 DOI: 10.1667/rade-23-00132.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 12/15/2023] [Indexed: 02/07/2024]
Abstract
The purpose of this investigation was to characterize the natural history of a murine total-abdominal-irradiation exposure model to measure gastrointestinal acute radiation injury. Male CD2F1 mice at 12 to 15 weeks old received total-abdominal irradiation using 4-MV linear accelerator X-rays doses of 0, 11, 13.5, 15, 15.75 and 16.5 Gy (2.75 Gy/min). Daily cage-side (i.e., in the animal housing room) observations of clinical signs and symptoms including body weights on all animals were measured up to 10 days after exposure. Jejunum tissues from cohorts of mice were collected at 1, 3, 7 and 10 days after exposure and radiation injury was assessed by histopathological analyses. Results showed time- and dose-dependent loss of body weight [for example at 7 days: 0.66 (±0.80) % loss for 0 Gy, 6.40 (±0.76) % loss at 11 Gy, 9.43 (±2.06) % loss at 13.5 Gy, 23.53 (± 1.91) % loss at 15 Gy, 29.97 (±1.16) % loss at 15.75 Gy, and 31.79 (±0.76) % loss at 16.5 Gy]. Negligible clinical signs and symptoms, except body weight changes, of radiation injury were observed up to 10 days after irradiation with doses of 11 to 15 Gy. Progressive increases in the severity of clinical signs and symptoms were found after irradiation with doses >15 Gy. Jejunum histology showed a progressive dose-dependent increase in injury. For example, at 7 days postirradiation, the percent of crypts, compared to controls, decreased to 82.3 (±9.5), 69.2 (±12.3), 45.4 (±11.9), 18.0 (±3.4), and 11.5 (± 1.8) with increases in doses from 11 to 16.5 Gy. A mucosal injury scoring system was used that mainly focused on changes in villus morphology damage (i.e., subepithelial spaces near the tips of the villi with capillary congestion, significant epithelial lifting along the length of the villi with a few denuded villus tips). Peak levels of total-abdominal irradiation induced effects on the mucosal injury score were seen 7 days after irradiation for doses ≥15 Gy, with a trend to show a decline after 7 days. A murine multiple-parameter gastrointestinal acute-radiation syndrome severity-scoring system was established based on clinical signs and symptoms that included measures of appearance (i.e., hunched and/or fluffed fur), respiratory rate, general (i.e., decreased mobility) and provoked behavior (i.e., subdued response to stimulation), weight loss, and feces/diarrhea score combined with jejunum mucosal-injury grade score. In summary, the natural-history radio-response for murine partial-body irradiation exposures is important for establishing a well-characterized radiation model system; here we established a multiple-parameter gastrointestinal acute-radiation syndrome severity-scoring system that provides a radiation injury gastrointestinal tissue-based assessment utility.
Collapse
Affiliation(s)
- David L Bolduc
- Scientific Research Department, Armed Forces Radiobiology Research Institute
| | - Lynnette H Cary
- Scientific Research Department, Armed Forces Radiobiology Research Institute
- Pharmacology and Molecular Therapeutics
| | - Juliann G Kiang
- Scientific Research Department, Armed Forces Radiobiology Research Institute
- Pharmacology and Molecular Therapeutics
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Lalitha Kurada
- Scientific Research Department, Armed Forces Radiobiology Research Institute
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Rockville, Maryland
| | - Vidya P Kumar
- Scientific Research Department, Armed Forces Radiobiology Research Institute
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Rockville, Maryland
| | - Sunshine A Edma
- Scientific Research Department, Armed Forces Radiobiology Research Institute
| | - Matthew G Olson
- Scientific Research Department, Armed Forces Radiobiology Research Institute
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Rockville, Maryland
| | - Vernieda B Vergara
- Scientific Research Department, Armed Forces Radiobiology Research Institute
| | - Dalton D Bistline
- Scientific Research Department, Armed Forces Radiobiology Research Institute
| | - Mario Reese
- Scientific Research Department, Armed Forces Radiobiology Research Institute
| | - Doreswamy Kenchegowda
- Scientific Research Department, Armed Forces Radiobiology Research Institute
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Rockville, Maryland
| | - Maureen Hood
- Biomedical Research Imaging Core at Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Department of Radiology & Radiological Sciences, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Alexandru Korotcov
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Rockville, Maryland
- Biomedical Research Imaging Core at Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Department of Radiology & Radiological Sciences, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Shalini Jaiswal
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Rockville, Maryland
- Biomedical Research Imaging Core at Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Department of Radiology & Radiological Sciences, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - William F Blakely
- Scientific Research Department, Armed Forces Radiobiology Research Institute
- Preventive Medicine and Statistics, Uniformed Services of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
7
|
Kiang JG, Cannon G, Olson MG, Zhai M, Woods AK, Xu F, Lin B, Li X, Hull L, Jiang S, Xiao M. Ciprofloxacin and pegylated G-CSF combined therapy mitigates brain hemorrhage and mortality induced by ionizing irradiation. Front Public Health 2023; 11:1268325. [PMID: 38162617 PMCID: PMC10756649 DOI: 10.3389/fpubh.2023.1268325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/14/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction Brain hemorrhage was found between 13 and 16 days after acute whole-body 9.5 Gy 60Co-γ irradiation (IR). This study tested countermeasures mitigating brain hemorrhage and increasing survival from IR. Previously, we found that pegylated G-CSF therapy (PEG) (i.e., Neulasta®, an FDA-approved drug) improved survival post-IR by 20-40%. This study investigated whether Ciprofloxacin (CIP) could enhance PEG-induced survival and whether IR-induced brain hemorrhage could be mitigated by PEG alone or combined with CIP. Methods B6D2F1 female mice were exposed to 60Co-γ-radiation. CIP was fed to mice for 21 days. PEG was injected on days 1, 8, and 15. 30-day survival and weight loss were studied in mice treated with vehicles, CIP, PEG, or PEG + CIP. For the early time point study, blood and sternums on days 2, 4, 9, and 15 and brains on day 15 post-IR were collected. Platelet numbers, brain hemorrhage, and histopathology were analyzed. The cerebellum/pons/medulla oblongata were detected with glial fibrillary acidic protein (GFAP), p53, p16, interleukin-18 (IL-18), ICAM1, Claudin 2, ZO-1, and complement protein 3 (C3). Results CIP + PEG enhanced survival after IR by 85% vs. the 30% improvement by PEG alone. IR depleted platelets, which was mitigated by PEG or CIP + PEG. Brain hemorrhage, both surface and intracranial, was observed, whereas the sham mice displayed no hemorrhage. CIP or CIP + PEG significantly mitigated brain hemorrhage. IR reduced GFAP levels that were recovered by CIP or CIP + PEG, but not by PEG alone. IR increased IL-18 levels on day 4 only, which was inhibited by CIP alone, PEG alone, or PEG + CIP. IR increased C3 on day 4 and day 15 and that coincided with the occurrence of brain hemorrhage on day 15. IR increased phosphorylated p53 and p53 levels, which was mitigated by CIP, PEG or PEG + CIP. P16, Claudin 2, and ZO-1 were not altered; ICAM1 was increased. Discussion CIP + PEG enhanced survival post-IR more than PEG alone. The Concurrence of brain hemorrhage, C3 increases and p53 activation post-IR suggests their involvement in the IR-induced brain impairment. CIP + PEG effectively mitigated the brain lesions, suggesting effectiveness of CIP + PEG therapy for treating the IR-induced brain hemorrhage by recovering GFAP and platelets and reducing C3 and p53.
Collapse
Affiliation(s)
- Juliann G. Kiang
- Radiation Combined Injury Program, Department of Scientific Research, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Georgetta Cannon
- Radiation Combined Injury Program, Department of Scientific Research, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
| | - Matthew G. Olson
- Radiation Combined Injury Program, Department of Scientific Research, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
| | - Min Zhai
- Radiation Combined Injury Program, Department of Scientific Research, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
| | - Akeylah K. Woods
- Radiation Combined Injury Program, Department of Scientific Research, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
| | - Feng Xu
- Radiation Combined Injury Program, Department of Scientific Research, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
| | - Bin Lin
- Radiation Combined Injury Program, Department of Scientific Research, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
| | - Xianghong Li
- Radiation Combined Injury Program, Department of Scientific Research, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
| | - Lisa Hull
- Radiation Combined Injury Program, Department of Scientific Research, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
| | - Suping Jiang
- Radiation Combined Injury Program, Department of Scientific Research, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
| | - Mang Xiao
- Radiation Combined Injury Program, Department of Scientific Research, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
| |
Collapse
|
8
|
Lu Q, Liang Y, Tian S, Jin J, Zhao Y, Fan H. Radiation-Induced Intestinal Injury: Injury Mechanism and Potential Treatment Strategies. TOXICS 2023; 11:1011. [PMID: 38133412 PMCID: PMC10747544 DOI: 10.3390/toxics11121011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/01/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023]
Abstract
Radiation-induced intestinal injury (RIII) is one of the most common intestinal complications caused by radiotherapy for pelvic and abdominal tumors and it seriously affects the quality of life of patients. However, the treatment of acute RIII is essentially symptomatic and nutritional support treatment and an ideal means of prevention and treatment is lacking. Researchers have conducted studies at the cellular and animal levels and found that some chemical or biological agents have good therapeutic effects on RIII and may be used as potential candidates for clinical treatment. This article reviews the injury mechanism and potential treatment strategies based on cellular and animal experiments to provide new ideas for the diagnosis and treatment of RIII in clinical settings.
Collapse
Affiliation(s)
- Qianying Lu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (Y.L.); (S.T.); (J.J.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Yangfan Liang
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (Y.L.); (S.T.); (J.J.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Sijia Tian
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (Y.L.); (S.T.); (J.J.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Jie Jin
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (Y.L.); (S.T.); (J.J.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Yanmei Zhao
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (Y.L.); (S.T.); (J.J.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Haojun Fan
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (Y.L.); (S.T.); (J.J.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| |
Collapse
|
9
|
Yang W, Li J, Hu J, Yuan X, Ding J, Jiang H, Wang G, Luo Q. Hypobaric hypoxia induces iron mobilization from liver and spleen and increases serum iron via activation of ghrelin/GHSR1a/MAPK signalling pathway in mice. Sci Rep 2023; 13:20254. [PMID: 37985861 PMCID: PMC10662372 DOI: 10.1038/s41598-023-47596-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 11/15/2023] [Indexed: 11/22/2023] Open
Abstract
Hypobaric hypoxia (HH) exposure affects appetite and serum iron levels in both humans and animals. Thus, whether appetite-regulating ghrelin is involved in iron regulation under HH needs to be elucidated. In vivo, C57BL/6J mice were placed in a hypobaric chamber to establish a 6000-m-high altitude exposure animal model. In vitro, mouse primary hepatocytes and peritoneal macrophages were exposed to hypoxia (1% O2) to examine the effects of ghrelin on iron-regulating proteins. HH obviously reduced the body weight of mice and significantly increased the levels of erythrocytes, and also significantly enhanced the levels of serum iron and plasma ghrelin. However, iron content in the liver and spleen was decreased, while ferroportin (Fpn) expression was increased. Moreover, ghrelin significantly induced Fpn and pERK expression in both hepatocytes and macrophages under hypoxia, which were reversed by pretreatment with growth hormone secretagogue receptor 1a (GHSR1a) antagonist or pERK inhibitor. Our findings indicated that HH leads to decreased appetite and insufficient dietary intake, which may negatively regulate the levels of ghrelin. Furthermore, GHSR1a/ERK signalling pathway is further activated to upregulate the expression of Fpn, and then promoting iron mobilization both in the liver/hepatocytes and spleen/macrophages in mice. Thus, these results revealed that ghrelin may be a potential iron regulatory hormone, and raised the possibility of ghrelin as a promising therapeutic target against iron disorders under HH.
Collapse
Affiliation(s)
- Wanping Yang
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine, Nantong University, 9 Seyuan Road, Chongchuan District, Nantong, 226019, Jiangsu, China
| | - Jiayan Li
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine, Nantong University, 9 Seyuan Road, Chongchuan District, Nantong, 226019, Jiangsu, China
| | - Jianan Hu
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine, Nantong University, 9 Seyuan Road, Chongchuan District, Nantong, 226019, Jiangsu, China
| | - Xiaoyu Yuan
- Department of Emergency, Affiliated Hospital of Nantong University, 20 XiSi Road, Nantong, 226001, China
| | - Jie Ding
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine, Nantong University, 9 Seyuan Road, Chongchuan District, Nantong, 226019, Jiangsu, China
| | - Hui Jiang
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine, Nantong University, 9 Seyuan Road, Chongchuan District, Nantong, 226019, Jiangsu, China
| | - Guohua Wang
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine, Nantong University, 9 Seyuan Road, Chongchuan District, Nantong, 226019, Jiangsu, China.
| | - Qianqian Luo
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine, Nantong University, 9 Seyuan Road, Chongchuan District, Nantong, 226019, Jiangsu, China.
| |
Collapse
|
10
|
Dushime H, Moreno SG, Linard C, Adrait A, Couté Y, Peltzer J, Messiaen S, Torres C, Bensemmane L, Lewandowski D, Romeo PH, Petit V, Gault N. Fetal Muse-based therapy prevents lethal radio-induced gastrointestinal syndrome by intestinal regeneration. Stem Cell Res Ther 2023; 14:201. [PMID: 37568164 PMCID: PMC10416451 DOI: 10.1186/s13287-023-03425-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Human multilineage-differentiating stress enduring (Muse) cells are nontumorigenic endogenous pluripotent-like stem cells that can be easily obtained from various adult or fetal tissues. Regenerative effects of Muse cells have been shown in some disease models. Muse cells specifically home in damaged tissues where they exert pleiotropic effects. Exposition of the small intestine to high doses of irradiation (IR) delivered after radiotherapy or nuclear accident results in a lethal gastrointestinal syndrome (GIS) characterized by acute loss of intestinal stem cells, impaired epithelial regeneration and subsequent loss of the mucosal barrier resulting in sepsis and death. To date, there is no effective medical treatment for GIS. Here, we investigate whether Muse cells can prevent lethal GIS and study how they act on intestinal stem cell microenvironment to promote intestinal regeneration. METHODS Human Muse cells from Wharton's jelly matrix of umbilical cord (WJ-Muse) were sorted by flow cytometry using the SSEA-3 marker, characterized and compared to bone-marrow derived Muse cells (BM-Muse). Under gas anesthesia, GIS mice were treated or not through an intravenous retro-orbital injection of 50,000 WJ-Muse, freshly isolated or cryopreserved, shortly after an 18 Gy-abdominal IR. No immunosuppressant was delivered to the mice. Mice were euthanized either 24 h post-IR to assess early small intestine tissue response, or 7 days post-IR to assess any regenerative response. Mouse survival, histological stainings, apoptosis and cell proliferation were studied and measurement of cytokines, recruitment of immune cells and barrier functional assay were performed. RESULTS Injection of WJ-Muse shortly after abdominal IR highly improved mouse survival as a result of a rapid regeneration of intestinal epithelium with the rescue of the impaired epithelial barrier. In small intestine of Muse-treated mice, an early enhanced secretion of IL-6 and MCP-1 cytokines was observed associated with (1) recruitment of monocytes/M2-like macrophages and (2) proliferation of Paneth cells through activation of the IL-6/Stat3 pathway. CONCLUSION Our findings indicate that a single injection of a small quantity of WJ-Muse may be a new and easy therapeutic strategy for treating lethal GIS.
Collapse
Affiliation(s)
- Honorine Dushime
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire Réparation et Transcription dans les cellules Souches (LRTS), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), CEA, 92260, Fontenay-aux-Roses, France
- Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, LRTS/iRCM/IBFJ, CEA, 92260, Fontenay-aux-Roses, France
| | - Stéphanie G Moreno
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire Réparation et Transcription dans les cellules Souches (LRTS), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), CEA, 92260, Fontenay-aux-Roses, France
- Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, LRTS/iRCM/IBFJ, CEA, 92260, Fontenay-aux-Roses, France
| | - Christine Linard
- Laboratory of Medical Radiobiology, Institute of Radiological Protection and Nuclear Safety, Fontenay-aux-Roses, France
| | - Annie Adrait
- Université Grenoble Alpes, Inserm, CEA, UMR BioSanté U1292, CNRS, FR2048, CEA, 38000, Grenoble, France
| | - Yohann Couté
- Université Grenoble Alpes, Inserm, CEA, UMR BioSanté U1292, CNRS, FR2048, CEA, 38000, Grenoble, France
| | - Juliette Peltzer
- Institut de Recherche Biomédicale des Armées (IRBA), 92141, Clamart, France
- UMR-S-MD 1197, Ministère des Armées et Université Paris Saclay, Villejuif, France
| | - Sébastien Messiaen
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire Réparation et Transcription dans les cellules Souches (LRTS), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), CEA, 92260, Fontenay-aux-Roses, France
- Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, LRTS/iRCM/IBFJ, CEA, 92260, Fontenay-aux-Roses, France
| | - Claire Torres
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire Réparation et Transcription dans les cellules Souches (LRTS), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), CEA, 92260, Fontenay-aux-Roses, France
- Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, LRTS/iRCM/IBFJ, CEA, 92260, Fontenay-aux-Roses, France
| | - Lydia Bensemmane
- Laboratory of Medical Radiobiology, Institute of Radiological Protection and Nuclear Safety, Fontenay-aux-Roses, France
| | - Daniel Lewandowski
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire Réparation et Transcription dans les cellules Souches (LRTS), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), CEA, 92260, Fontenay-aux-Roses, France
- Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, LRTS/iRCM/IBFJ, CEA, 92260, Fontenay-aux-Roses, France
| | - Paul-Henri Romeo
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire Réparation et Transcription dans les cellules Souches (LRTS), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), CEA, 92260, Fontenay-aux-Roses, France
- Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, LRTS/iRCM/IBFJ, CEA, 92260, Fontenay-aux-Roses, France
| | - Vanessa Petit
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire Réparation et Transcription dans les cellules Souches (LRTS), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), CEA, 92260, Fontenay-aux-Roses, France.
- Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, LRTS/iRCM/IBFJ, CEA, 92260, Fontenay-aux-Roses, France.
| | - Nathalie Gault
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire Réparation et Transcription dans les cellules Souches (LRTS), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), CEA, 92260, Fontenay-aux-Roses, France.
- Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, LRTS/iRCM/IBFJ, CEA, 92260, Fontenay-aux-Roses, France.
| |
Collapse
|
11
|
Yin J, Xie X, Yao J, Jin X, Jiang H, Ji C. Transcription factor Krüppel-like factor 4 upregulated G protein-coupled receptor 30 alleviates intestinal inflammation and apoptosis, and protects intestinal integrity from intestinal ischemia-reperfusion injury. Immun Inflamm Dis 2023; 11:e940. [PMID: 37506161 PMCID: PMC10373568 DOI: 10.1002/iid3.940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 06/24/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
INTRODUCTION Intestinal ischemia/reperfusion (I/R) injury is a common clinical event occurring during multiple clinical pathological processes. Here, we designed this paper to discuss the role of G protein-coupled receptor 30 (GPR30) playing in intestinal I/R injury. METHODS An oxygen-glucose deprivation/reoxygenation (OGD/R) cell model was established to simulate the pathological process of I/R injury. With the application of enzyme-linked immunosorbent assay, TUNEL, and transepithelial electrical resistance (TEER) assays, the levels of inflammatory cytokines, cell apoptosis, and intestinal integrity were estimated. The corresponding proteins were estimated by applying western blot. Immunofluorescence was conducted to examine N-terminal Gasdermin D (GSDMD-N) expression. The interplay between KLF4 and GPR30 was demonstrated by dual-luciferase reporter assay and chromatin immunoprecipitation. RESULTS The results showed that GPR30 was downregulated in Caco-2 cells exposed to OGD/R. GPR30 overexpression reduced the production of TNF-α, IL-6, IL-1β, and IL-18, the TUNEL-positive cells, as well as the contents of p-p65, Cox-2, Inos, Bax, and cleaved-PARP, but elevated the expression of Bcl-2 in OGD/R-induced Caco-2 cells. In addition, OGD/R-induced the reduction of TEER value and reduced expression of tight junction proteins in Caco-2 cells, which was partially restored by GPR30 overexpression. Furthermore, GPR30 suppressed nod-like receptor pyrin 3 inflammasome and GSDMD-N expression. It was evidenced that Krüppel-like factor 4 (KLF4) could directly bind to GPR30 promoter and positively regulate GPR30 expression. The regulation of GPR30 overexpression above was weakened by KLF4 knockdown. CONCLUSION Collectively, our findings suggested that KLF4 could transcriptionally upregulate GPR30, and GPR30 prevented intestine I/R injury by inhibiting inflammation and apoptosis, and maintaining intestinal integrity that provides potential targets for mitigating the I/R injury.
Collapse
Affiliation(s)
- Jie Yin
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaoli Xie
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jinfeng Yao
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaoxu Jin
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Huiqing Jiang
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chenguang Ji
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
12
|
Chen L, Wang Z, Wu J, Yao Q, Peng J, Zhang C, Chen H, Li Y, Jiang Z, Liu Y, Shi C. Released dsDNA-triggered inflammasomes serve as intestinal radioprotective targets. Clin Transl Immunology 2023; 12:e1452. [PMID: 37333051 PMCID: PMC10276537 DOI: 10.1002/cti2.1452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 05/05/2023] [Accepted: 05/18/2023] [Indexed: 06/20/2023] Open
Abstract
Objectives Intestinal mucositis is the major side effect during abdominal or pelvic radiotherapy, but the underlying immunogen remains to be further characterised and few radioprotective agents are available. This study investigated the role of dsDNA-triggered inflammasomes in intestinal mucositis during radiotherapy. Methods Pro-inflammatory cytokines were detected by ELISA. Radiation-induced intestinal injury in mice was analyzed by means of survival curves, body weight, HE staining of intestines, and intestinal barrier integrity. Western blot, immunofluorescence staining, co-immunoprecipitation assay and flow cytometry were used to investigate the regulatory role of dsDNA on inflammasomes. Results Here, we show that a high level of IL-1β and IL-18 is associated with diarrhoea in colorectal cancer (CRC) patients during radiotherapy, which accounts for intestinal radiotoxicity. Subsequently, we found that the dose-dependently released dsDNA from the intestinal epithelial cells (IECs) serves as the potential immunogenic molecule for radiation-induced intestinal mucositis. Our results further indicate that the released dsDNA transfers into the macrophages in an HMGB1/RAGE-dependent manner and then triggers absent in melanoma 2 (AIM2) inflammasome activation and the IL-1β and IL-18 secretion. Finally, we show that the FDA-approved disulfiram (DSF), a newly identified inflammasome inhibitor, could mitigate intestinal radiotoxicity by controlling inflammasome. Conclusion These findings indicate that the extracellular self-dsDNA released from the irradiated IECs is a potential immunogen to stimulate immune cells and trigger the subsequent intestinal mucositis, while blunting the dsDNA-triggered inflammasome in macrophages may represent an exciting therapeutic strategy for side effects control during abdominal radiotherapy.
Collapse
Affiliation(s)
- Long Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force MedicineArmy Medical UniversityChongqingChina
- Shigatse Branch, Xinqiao Hospital, Army 953 HospitalArmy Medical UniversityShigatseChina
| | - Ziwen Wang
- Department of CardiologyGeriatric Cardiovascular Disease Research and Treatment Center, 252 Hospital of PLABaodingChina
| | - Jie Wu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force MedicineArmy Medical UniversityChongqingChina
| | - Quan Yao
- Integrative Cancer Center & Cancer Clinical Research Center, Sichuan Cancer Center, School of Medicine, Sichuan Cancer Hospital & InstituteUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Jingjing Peng
- Department of OncologyWestern Theater General HospitalChengduChina
| | - Chi Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force MedicineArmy Medical UniversityChongqingChina
| | - Hongdan Chen
- Breast and Thyroid Surgical Department, Chongqing General HospitalUniversity of Chinese Academy of SciencesChongqingChina
| | - Yingjie Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force MedicineArmy Medical UniversityChongqingChina
| | - Zhongyong Jiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force MedicineArmy Medical UniversityChongqingChina
| | - Yunsheng Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force MedicineArmy Medical UniversityChongqingChina
| | - Chunmeng Shi
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force MedicineArmy Medical UniversityChongqingChina
| |
Collapse
|
13
|
Kiang JG, Blakely WF. Combined radiation injury and its impacts on radiation countermeasures and biodosimetry. Int J Radiat Biol 2023; 99:1055-1065. [PMID: 36947602 PMCID: PMC10947598 DOI: 10.1080/09553002.2023.2188933] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/10/2023] [Accepted: 03/01/2023] [Indexed: 03/24/2023]
Abstract
PURPOSE Preparedness for medical responses to major radiation accidents and the increasing threat of nuclear warfare worldwide necessitates an understanding of the complexity of combined radiation injury (CI) and identifying drugs to treat CI is inevitably critical. The vital sign and survival after CI were presented. The molecular mechanisms, such as microRNA pathways, NF-κB-iNOS-IL-18 pathway, C3 production, the AKT-MAPK cross-talk, and TLR/MMP increases, underlying CI in relation to organ injury and mortality were analyzed. At present, no FDA-approved drug to protect, mitigate, or treat CI is available. The development of CI-specific medical countermeasures was reviewed. Because of the worsened acute radiation syndrome resulting from CI, diagnostic triage can be problematic. Therefore, biodosimetry and CI are bundled together with the need to establish effective triage methods with CI. CONCLUSIONS CI mouse model studies at AFRRI are reviewed addressing molecular responses, findings from medical countermeasures, and a proposed plasma proteomic biodosimetry approach based on a panel of radiation-responsive biomarkers (i.e., CD27, Flt-3L, GM-CSF, CD45, IL-12, TPO) negligibly influenced by wounding in an algorithm used for dose predictions is described.
Collapse
Affiliation(s)
- Juliann G. Kiang
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - William F. Blakely
- Biodosimetry Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
14
|
Fang Z, Lv Y, Zhang H, He Y, Gao H, Chen C, Wang D, Chen P, Tang S, Li J, Qiu Z, Shi X, Chen L, Yang J, Chen X. A multifunctional hydrogel loaded with two nanoagents improves the pathological microenvironment associated with radiation combined with skin wounds. Acta Biomater 2023; 159:111-127. [PMID: 36736645 DOI: 10.1016/j.actbio.2023.01.052] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/02/2023] [Accepted: 01/20/2023] [Indexed: 02/04/2023]
Abstract
Persistent oxidative stress and recurring waves of inflammation with excessive reactive oxygen species (ROS) and free radical accumulation could be generated by radiation. Exposure to radiation in combination with physical injuries such as wound trauma would produce a more harmful set of medical complications, which was known as radiation combined with skin wounds (RCSWs). However, little attention has been given to RCSW research despite the unsatisfactory therapeutic outcomes. In this study, a dual-nanoagent-loaded multifunctional hydrogel was fabricated to ameliorate the pathological microenvironment associated with RCSWs. The injectable, adhesive, and self-healing hydrogel was prepared by crosslinking carbohydrazide-modified gelatin (Gel-CDH) and oxidized hyaluronic acid (OHA) through the Schiff-base reaction under mild condition. Polydopamine nanoparticles (PDA-NPs) and mesenchymal stem cell-secreted small extracellular vesicles (MSC-sEV) were loaded to relieve radiation-produced tissue inflammation and oxidation impairment and enhance cell vitality and angiogenesis individually or jointly. The proposed PDA-NPs@MSC-sEV hydrogel enhanced cell vitality, as shown by cell proliferation, migration, colony formation, and cell cycle and apoptosis assays in vitro, and promoted reepithelization by attenuating microenvironment pathology in vivo. Notably, a gene set enrichment analysis of proteomic data revealed significant enrichment with adipogenic and hypoxic pathways, which play prominent roles in wound repair. Specifically, target genes were predicted based on differential transcription factor expression. The results suggested that MSC-sEV- and PDA-NP-loaded multifunctional hydrogels may be promising nanotherapies for RCSWs. STATEMENT OF SIGNIFICANCE: The small extracellular vesicle (sEV) has distinct advantages compared with MSCs, and polydopamine nanoparticles (PDA-NPs), known as the biological materials with good cell affinity and histocompatibility which have been reported to scavenge ROS free radicals. In this study, an adhesive, injectable, self-healing, antibacterial, ROS scavenging and amelioration of the radiation related microenvironment hydrogel encapsulating nanoscale particles of MSC-sEV and PDA-NPs (PDA-NPs@MSC-sEV hydrogel) was synthesized for promoting radiation combined with skin wounds (RCSWs). GSEA analysis profiled by proteomics data revealed significant enrichments in the regulations of adipogenic and hypoxic pathways with this multi-functional hydrogel. This is the first report of combining this two promising nanoscale agents for the special skin wounds associated with radiation.
Collapse
Affiliation(s)
- Zhuoqun Fang
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou 350001, China; Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou 350001, China; Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, 350001, China; Department of Stem Cell Research Institute, Fujian Medical University, Fuzhou 350004, China
| | - Yicheng Lv
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China; Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Haoruo Zhang
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou 350001, China; Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou 350001, China; Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, 350001, China; Department of Stem Cell Research Institute, Fujian Medical University, Fuzhou 350004, China
| | - Yuxiang He
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China; Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Hangqi Gao
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou 350001, China; Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou 350001, China; Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, 350001, China; Department of Stem Cell Research Institute, Fujian Medical University, Fuzhou 350004, China
| | - Caixiang Chen
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou 350001, China; Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou 350001, China; Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, 350001, China; Department of Stem Cell Research Institute, Fujian Medical University, Fuzhou 350004, China
| | - Dezhi Wang
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou 350001, China; Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou 350001, China; Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, 350001, China; Department of Stem Cell Research Institute, Fujian Medical University, Fuzhou 350004, China
| | - Penghong Chen
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou 350001, China; Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou 350001, China; Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, 350001, China; Department of Stem Cell Research Institute, Fujian Medical University, Fuzhou 350004, China
| | - Shijie Tang
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou 350001, China; Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou 350001, China; Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, 350001, China; Department of Stem Cell Research Institute, Fujian Medical University, Fuzhou 350004, China
| | - Junjing Li
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou 350001, China; Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou 350001, China; Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, 350001, China; Department of Breast Surgery, Quanzhou First Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Zhihuang Qiu
- Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, 350001, China; Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Xian'ai Shi
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China; Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Liangwan Chen
- Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, 350001, China; Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China.
| | - Jianmin Yang
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China; Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China.
| | - Xiaosong Chen
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou 350001, China; Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou 350001, China; Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, 350001, China.
| |
Collapse
|
15
|
Kaempferol Reverses Acute Kidney Injury in Septic Model by Inhibiting NF-κB/AKT Signaling Pathway. J Food Biochem 2023. [DOI: 10.1155/2023/1353449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Sepsis is the main cause of acute kidney injury (AKI), mainly due to systemic immune dysregulation. Kaempferol (KAE) is a natural flavonoid compound with multiple biological activities including anti-inflammatory, antioxidant, and antiapoptotic properties. In this study, we constructed a sepsis-induced AKI mouse model and an LPS-induced glomerular mesangial cell (HK-2) in vitro sepsis AKI model. We found that KAE ameliorated sepsis-induced renal pathological damage, reversed renal function damage, and inhibited p-p65 and p-AKT protein expression. In addition, KAE reversed LPS-induced proliferation and inhibited apoptosis in HK-2 cells. These studies suggest that KAE reverses sepsis by inhibiting activation of the NF-κB/AKT pathway to reverse acute kidney injury.
Collapse
|
16
|
Wang L, Lin B, Zhai M, Cui W, Hull L, Zizzo A, Li X, Kiang JG, Xiao M. Deteriorative Effects of Radiation Injury Combined with Skin Wounding in a Mouse Model. TOXICS 2022; 10:toxics10120785. [PMID: 36548618 PMCID: PMC9783596 DOI: 10.3390/toxics10120785] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/07/2022] [Accepted: 12/11/2022] [Indexed: 05/14/2023]
Abstract
Radiation-combined injury (RCI) augments the risk of morbidity and mortality when compared to radiation injury (RI) alone. No FDA-approved medical countermeasures (MCMs) are available for treating RCI. Previous studies implied that RI and RCI elicit differential mechanisms leading to their detrimental effects. We hypothesize that accelerating wound healing improves the survival of RCI mice. In the current study, we examined the effects of RCI at different doses on lethality, weight loss, wound closure delay, and proinflammatory status, and assessed the relative contribution of systemic and local elements to their delayed wound closure. Our data demonstrated that RCI increased the lethality and weight loss, delayed skin wound closure, and induced a systemic proinflammatory status in a radiation dose-dependent manner. We also demonstrated that delayed wound closure did not specifically depend on the extent of hematopoietic suppression, but was significantly influenced by the toxicity of the radiation-induced systemic inflammation and local elements, including the altered levels of proinflammatory chemokines and factors, and the dysregulated collagen homeostasis in the wounded area. In conclusion, the results from our study indicate a close association between delayed wound healing and the significantly altered pathways in RCI mice. This insightful information may contribute to the evaluation of the prognosis of RCI and development of MCMs for RCI.
Collapse
Affiliation(s)
- Li Wang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Bin Lin
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Min Zhai
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Wanchang Cui
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Lisa Hull
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Alex Zizzo
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Xianghong Li
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Juliann G. Kiang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Mang Xiao
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Correspondence: ; Tel.: +1-301-295-2597
| |
Collapse
|
17
|
Jiang M, Wan S, Dai X, Ye Y, Hua W, Ma G, Pang X, Wang H, Shi B. Protective effect of ghrelin on intestinal I/R injury in rats. Open Med (Wars) 2022; 17:1308-1317. [PMID: 35937002 PMCID: PMC9307145 DOI: 10.1515/med-2022-0520] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 06/14/2022] [Accepted: 06/15/2022] [Indexed: 11/15/2022] Open
Abstract
Abstract
This study aimed to investigate whether ghrelin affected the autophagy and inflammatory response of intestinal intraepithelial lymphocytes (IELs) by regulating the NOD2/Beclin-1 pathway in an intestinal ischemia–reperfusion (I/R) injury model. Twenty hours after implementing the intestinal I/R injury rat model, the small intestine and both lungs were collected for histological analysis. The morphological changes in the intestinal mucosa epithelium and lung tissues were evaluated using hematoxylin-eosin staining. The activity of autophagic vacuoles and organ injury were evaluated using electron microscopy. The cytokine levels (IL-10 and TNF-α) in IEL cells and lung tissue were determined using enzyme-linked immunosorbent assay. RT-qPCR and western blot assays were conducted to check the NOD2, Beclin-1, and ATG16 levels. Ghrelin relieved the I/R-induced destruction of the intestinal mucosa epithelium and lung tissues. Moreover, ghrelin enhanced autophagy in the intestinal epithelium and lungs of I/R rats. In addition, the levels of autophagy-associated proteins (Beclin-1, ATG16, and NOD2) were higher in the ghrelin treatment group than in rats with I/R. Ghrelin reduced significantly the IL-10 and TNF-α levels. However, these changes were reversed by the NOD2 antagonist. In conclusion, ghrelin may relieve I/R-induced acute intestinal mucosal damage, autophagy disorder, and inflammatory response in IELs by regulating the NOD2/Beclin-1 pathway.
Collapse
Affiliation(s)
- Meng Jiang
- Department of Emergency Intensive Care Unit, Yangpu Hospital, Tongji University , Shanghai 200090 , China
| | - Shengxia Wan
- Department of Neurology, The Fourth Affiliated Hospital of Jiangsu University , Zhenjiang 212000 , China
| | - Xiaoyong Dai
- Department of Emergency Intensive Care Unit, Yangpu Hospital, Tongji University , Shanghai 200090 , China
| | - Youwen Ye
- Department of Emergency Intensive Care Unit, Yangpu Hospital, Tongji University , Shanghai 200090 , China
| | - Wei Hua
- Department of Emergency Intensive Care Unit, Yangpu Hospital, Tongji University , Shanghai 200090 , China
| | - Guoguang Ma
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University , Shanghai 200032 , China
| | - Xiufeng Pang
- Department of Emergency Intensive Care Unit, Yangpu Hospital, Tongji University , Shanghai 200090 , China
| | - Huanhuan Wang
- Department of Emergency Intensive Care Unit, Yangpu Hospital, Tongji University , Shanghai 200090 , China
| | - Bin Shi
- Department of Emergency Intensive Care Unit, Yangpu Hospital, Tongji University , Shanghai 200090 , China
| |
Collapse
|
18
|
Kiang JG, Cannon G, Olson MG, Smith JT, Anderson MN, Zhai M, Umali MV, Ho K, Ho C, Cui W, Xiao M. Female Mice are More Resistant to the Mixed-Field (67% Neutron + 33% Gamma) Radiation-Induced Injury in Bone Marrow and Small Intestine than Male Mice due to Sustained Increases in G-CSF and the Bcl-2/Bax Ratio and Lower miR-34a and MAPK Activation. Radiat Res 2022; 198:120-133. [PMID: 35452510 DOI: 10.1667/rade-21-00201.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 04/04/2022] [Indexed: 11/03/2022]
Abstract
In nuclear and radiological incidents, overexposure to ionizing radiation is life-threatening. It is evident that radiation depletes blood cells and increases circulating cytokine/chemokine concentrations as well as mortality. While microglia cells of female mice have been observed to be less damaged by radiation than in male mice, it is unclear whether sex affects physio-pathological responses in the bone marrow (BM) and gastrointestinal system (GI). We exposed B6D2F1 male and female mice to 0, 1.5, 3, or 6 Gy with mixed-field radiation containing 67% neutron and 33% gamma at a dose rate of 0.6 Gy/min. Blood and tissues were collected on days 1, 4, and 7 postirradiation. Radiation increased cytokines/chemokines in the femurs and ilea of female and male mice in a dose-dependent manner. Cytokines and chemokines reached a peak on day 4 and declined on day 7 with the exception of G-CSF which continued to increase on day 7 in female mice but not in male mice. MiR-34a (a Bcl-2 inhibitor), G-CSF (a miR-34a inhibitor), MAPK activation (pro-cell death), and citrulline (a biomarker of entro-epithelial proliferation), active caspase-3 (a biomarker of apoptosis) and caspase-1activated gasdermin D (a pyroptosis biomarker) were measured in the sternum, femur BM and ileum. Sternum histopathology analysis with H&E staining and femur BM cell counts as well as Flt-3L showed that BM cellularity was not as diminished in females, with males showing a 50% greater decline on day 7 postirradiation, mainly mediated by pyroptosis as indicated by increased gasdermin D in femur BM samples. Ileum injury, such as villus height and crypt depth, was also 43% and 30%, respectively, less damaged in females than in males. The severity of injury in both sexes was consistent with the citrulline and active caspase-3 measurements as well as active caspase-1 and gasdermin D measurements, suggesting apoptosis and pyroptosis occurred. On day 7, G-CSF in the ileum of female mice continued to be elevated by sevenfold, whereas G-CSF in the ileum of male mice returned to baseline. Furthermore, G-CSF is known to inhibit miR-34a expression, which in ileum on day 1 displayed a 3- to 4-fold increase in female mice after mixed-field (67% neutron + 33% gamma) irradiation, as compared to a 5- to 9-fold increase in male mice. Moreover, miR-34a blocked Bcl-2 expression. Mixed-field (60% neutron + 33% gamma) radiation induced more Bcl-2 in females than in males. On day 7, AKT activation was found in the ileums of females and males. However, MAPK activation including ERK, JNK, and p38 showed no changes in the ileum of females (by 0-fold; P > 0.05), whereas the MAPK activation was increased in the ileum of males (by 100-fold; P < 0.05). Taken together, the results suggest that organ injury from mixed-field (67% neutron + 33% gamma) radiation is less severe in females than in males, likely due to increased G-CSF, less MAPK activation, low miR-34a and increased Bcl-2/Bax ratio.
Collapse
Affiliation(s)
- Juliann G Kiang
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland.,Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland.,Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Georgetta Cannon
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Matthew G Olson
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Joan T Smith
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | | | - Min Zhai
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - M Victoria Umali
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Kevin Ho
- Department of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Connie Ho
- School of Medicine, University of California, Los Angeles, California
| | - Wanchang Cui
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Mang Xiao
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
19
|
Transcriptomics of Wet Skin Biopsies Predict Early Radiation-Induced Hematological Damage in a Mouse Model. Genes (Basel) 2022; 13:genes13030538. [PMID: 35328091 PMCID: PMC8952434 DOI: 10.3390/genes13030538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/28/2022] [Accepted: 03/16/2022] [Indexed: 12/04/2022] Open
Abstract
The lack of an easy and fast radiation-exposure testing method with a dosimetric ability complicates triage and treatment in response to a nuclear detonation, radioactive material release, or clandestine exposure. The potential of transcriptomics in radiation diagnosis and prognosis were assessed here using wet skin (blood/skin) biopsies obtained at hour 2 and days 4, 7, 21, and 28 from a mouse radiation model. Analysis of significantly differentially transcribed genes (SDTG; p ≤ 0.05 and FC ≥ 2) during the first post-exposure week identified the glycoprotein 6 (GP-VI) signaling, the dendritic cell maturation, and the intrinsic prothrombin activation pathways as the top modulated pathways with stable inactivation after lethal exposures (20 Gy) and intermittent activation after sublethal (1, 3, 6 Gy) exposure time points (TPs). Interestingly, these pathways were inactivated in the late TPs after sublethal exposure in concordance with a delayed deleterious effect. Modulated transcription of a variety of collagen types, laminin, and peptidase genes underlay the modulated functions of these hematologically important pathways. Several other SDTGs related to platelet and leukocyte development and functions were identified. These results outlined genetic determinants that were crucial to clinically documented radiation-induced hematological and skin damage with potential countermeasure applications.
Collapse
|
20
|
Chen ZY, Xiao HW, Dong JL, Li Y, Wang B, Fan SJ, Cui M. Gut Microbiota-Derived PGF2α Fights against Radiation-Induced Lung Toxicity through the MAPK/NF-κB Pathway. Antioxidants (Basel) 2021; 11:antiox11010065. [PMID: 35052569 PMCID: PMC8773112 DOI: 10.3390/antiox11010065] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/24/2021] [Accepted: 12/24/2021] [Indexed: 12/28/2022] Open
Abstract
Radiation pneumonia is a common and intractable side effect associated with radiotherapy for chest cancer and involves oxidative stress damage and inflammation, prematurely halting the remedy and reducing the life quality of patients. However, the therapeutic options for the complication have yielded disappointing results in clinical application. Here, we report an effective avenue for fighting against radiation pneumonia. Faecal microbiota transplantation (FMT) reduced radiation pneumonia, scavenged oxidative stress and improved lung function in mouse models. Local chest irradiation shifted the gut bacterial taxonomic proportions, which were preserved by FMT. The level of gut microbiota-derived PGF2α decreased following irradiation but increased after FMT. Experimental mice with PGF2α replenishment, via an oral route, exhibited accumulated PGF2α in faecal pellets, peripheral blood and lung tissues, resulting in the attenuation of inflammatory status of the lung and amelioration of lung respiratory function following local chest irradiation. PGF2α activated the FP/MAPK/NF-κB axis to promote cell proliferation and inhibit apoptosis with radiation challenge; silencing MAPK attenuated the protective effect of PGF2α on radiation-challenged lung cells. Together, our findings pave the way for the clinical treatment of radiotherapy-associated complications and underpin PGF2α as a gut microbiota-produced metabolite.
Collapse
Affiliation(s)
- Zhi-Yuan Chen
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300110, China; (Z.-Y.C.); (J.-L.D.); (Y.L.); (B.W.)
| | - Hui-Wen Xiao
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China;
| | - Jia-Li Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300110, China; (Z.-Y.C.); (J.-L.D.); (Y.L.); (B.W.)
| | - Yuan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300110, China; (Z.-Y.C.); (J.-L.D.); (Y.L.); (B.W.)
| | - Bin Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300110, China; (Z.-Y.C.); (J.-L.D.); (Y.L.); (B.W.)
| | - Sai-Jun Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300110, China; (Z.-Y.C.); (J.-L.D.); (Y.L.); (B.W.)
- Correspondence: (S.-J.F.); (M.C.)
| | - Ming Cui
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300110, China; (Z.-Y.C.); (J.-L.D.); (Y.L.); (B.W.)
- Correspondence: (S.-J.F.); (M.C.)
| |
Collapse
|
21
|
Zhao L, Qi X, Cai T, Fan Z, Wang H, Du X. Gelatin hydrogel/contact lens composites as rutin delivery systems for promoting corneal wound healing. Drug Deliv 2021; 28:1951-1961. [PMID: 34623206 PMCID: PMC8475096 DOI: 10.1080/10717544.2021.1979126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Corneal wound healing is a highly regulated biological process that is of importance for reducing the risk of blinding corneal infections and inflammations. Traditional eye drop was the main approach for promoting corneal wound healing. However, its low bioavailability required a high therapeutic concentration, which can lead to ocular or even systemic side effects. To develop a safe and effective method for treating corneal injury, we fabricated rutin-encapsulated gelatin hydrogel/contact lens composites by dual crosslinking reactions including in situ free radical polymerization and carboxymethyl cellulose/N-hydroxysulfosuccinimide crosslinking. In vitro drug release results evidenced that rutin in the composites could be sustainedly released for up to 14 days. In addition, biocompatibility assay indicated nontoxicity of the composites. Finally, the effect of rutin-encapsulated composites on the healing of the corneal injury in rabbits was investigated. The injury was basically cured in corneas using rutin-encapsulated composites (healing rate, 98.3% ± 0.7%) at 48 h post-operation, while the damage was still present in corneas using the composite (healing rate, 87.0% ± 4.5%). Further proteomics analysis revealed that corneal wound healing may be promoted by the ERK/MAPK and PI3K/AKT signal pathways. These results inform a potential intervention strategy to facilitate corneal wound healing in humans.
Collapse
Affiliation(s)
- Lianghui Zhao
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, Shandong, China.,State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, Shandong, China
| | - Xia Qi
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, Shandong, China.,State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, Shandong, China
| | - Tao Cai
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, Shandong, China.,State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, Shandong, China
| | - Zheng Fan
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, Shandong, China.,State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, Shandong, China
| | - Hongwei Wang
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, Shandong, China.,State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, Shandong, China
| | - Xianli Du
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, Shandong, China.,State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, Shandong, China
| |
Collapse
|
22
|
Taliaferro LP, Cassatt DR, Horta ZP, Satyamitra MM. Meeting Report: A Poly-Pharmacy Approach to Mitigate Acute Radiation Syndrome. Radiat Res 2021; 196:436-446. [PMID: 34237144 PMCID: PMC8532024 DOI: 10.1667/rade-21-00048.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 06/03/2021] [Indexed: 11/03/2022]
Abstract
The National Institute of Allergy and Infectious Diseases, Radiation and Nuclear Countermeasures Program, was tasked by the United States Congress and the U.S. Department of Health and Human Services to identify and fund early-to-mid-stage development of medical countermeasures (MCMs) to treat radiation-induced injuries. In developing MCMs to treat various sub-syndromes (e.g., hematopoietic, gastrointestinal, lung), it is important to investigate whether a poly-pharmacy approach (i.e., drug cocktails) can provide additive benefits to mitigate injuries arising from the acute radiation syndrome (ARS). In addition, potential drug-drug interactions must be examined. For this reason, a workshop was held, which centered on understanding the current state of research investigating poly-pharmacy approaches to treat radiation injuries. The first session set the stage with an introduction to the concept of operations or support available for the response to a nuclear incident, as this is the key to any emergency response, including MCM availability and distribution. The second session followed the natural history of ARS in both humans and animal models to underscore the complexity of ARS and why a poly-pharmacy approach may be necessary. The third session featured talks from investigators conducting current MCM poly-pharmacy research. The meeting closed with a focus on regulatory considerations for the development of poly-pharmacy approaches or combination treatments for ARS.
Collapse
Affiliation(s)
- Lanyn P. Taliaferro
- Radiation and Nuclear Countermeasures Program (RNCP), Division of
Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy
and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville,
Maryland
| | - David R. Cassatt
- Radiation and Nuclear Countermeasures Program (RNCP), Division of
Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy
and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville,
Maryland
| | | | - Merriline M. Satyamitra
- Radiation and Nuclear Countermeasures Program (RNCP), Division of
Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy
and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville,
Maryland
| |
Collapse
|
23
|
Wang L, Zhai M, Lin B, Cui W, Hull L, Li X, Anderson MN, Smith JT, Umali MV, Jiang S, Kiang JG, Xiao M. PEG-G-CSF and L-Citrulline Combination Therapy for Mitigating Skin Wound Combined Radiation Injury in a Mouse Model. Radiat Res 2021; 196:113-127. [PMID: 33914884 PMCID: PMC8344563 DOI: 10.1667/rade-20-00151.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 03/26/2021] [Indexed: 11/03/2022]
Abstract
Radiation combined injury (RCI, radiation exposure coupled with other forms of injury, such as burn, wound, hemorrhage, blast, trauma and/or sepsis) comprises approximately 65% of injuries from a nuclear explosion, and greatly increases the risk of morbidity and mortality when compared to that of radiation injury alone. To date, no U.S. Food and Drug Administration (FDA)-approved countermeasures are available for RCI. Currently, three leukocyte growth factors (Neupogen®, Neulasta® and Leukine®) have been approved by the FDA for mitigating the hematopoietic acute radiation syndrome. However these granulocyte-colony-stimulating factor (G-CSF) and granulocyte-macrophage colony-stimulating factor (GM-CSF) products have failed to increase 30-day survival of mice after RCI, suggesting a more complicated biological mechanism is in play for RCI than for radiation injury. In the current study, the mitigative efficacy of combination therapy using pegylated (PEG)-G-CSF (Neulasta) and -citrulline was evaluated in an RCI mouse model. L-citrulline is a neutral alpha-amino acid shown to improve vascular endothelial function in cardiovascular diseases. Three doses of PEG-G-CSF at 1 mg/kg, subcutaneously administered on days 1, 8 and 15 postirradiation, were supplemented with oral -citrulline (1 g/kg), once daily from day 1 to day 21 postirradiation. The combination treatment significantly improved the 30-day survival of mice after RCI from 15% (vehicle-treated) to 42%, and extended the median survival time by 4 days, as compared to vehicle controls. In addition, the combination therapy significantly increased body weight and bone marrow stem and progenitor cell clonogenicity in RCI mice, and accelerated recovery from RCI-induced intestinal injury, compared to animals treated with vehicle. Treatment with -citrulline alone also accelerated skin wound healing after RCI. In conclusion, these data indicate that the PEG-G-CSF and -citrulline combination therapy is a potentially effective countermeasure for mitigating RCI, likely by enhancing survival of the hematopoietic stem/progenitor cells and accelerating recovery from the RCI-induced intestinal injury and skin wounds.
Collapse
Affiliation(s)
- Li Wang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Min Zhai
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Bin Lin
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Wanchang Cui
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Lisa Hull
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Xianghong Li
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Marsha N. Anderson
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Joan T. Smith
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Maria Victoria Umali
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Suping Jiang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Juliann G. Kiang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
- Department of Pharmacology and Molecular Therapy, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Mang Xiao
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| |
Collapse
|
24
|
Gorbunov NV, Kiang JG. Brain Damage and Patterns of Neurovascular Disorder after Ionizing Irradiation. Complications in Radiotherapy and Radiation Combined Injury. Radiat Res 2021; 196:1-16. [PMID: 33979447 PMCID: PMC8297540 DOI: 10.1667/rade-20-00147.1] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 04/02/2021] [Indexed: 12/31/2022]
Abstract
Exposure to ionizing radiation, mechanical trauma, toxic chemicals or infections, or combinations thereof (i.e., combined injury) can induce organic injury to brain tissues, the structural disarrangement of interactive networks of neurovascular and glial cells, as well as on arrays of the paracrine and systemic destruction. This leads to subsequent decline in cognitive capacity and decompensation of mental health. There is an ongoing need for improvement in mitigating and treating radiation- or combined injury-induced brain injury. Cranial irradiation per se can cause a multifactorial encephalopathy that occurs in a radiation dose- and time-dependent manner due to differences in radiosensitivity among the various constituents of brain parenchyma and vasculature. Of particular concern are the radiosensitivity and inflammation susceptibility of: 1. the neurogenic and oligodendrogenic niches in the subependymal and hippocampal domains; and 2. the microvascular endothelium. Thus, cranial or total-body irradiation can cause a plethora of biochemical and cellular disorders in brain tissues, including: 1. decline in neurogenesis and oligodendrogenesis; 2. impairment of the blood-brain barrier; and 3. ablation of vascular capillary. These changes, along with cerebrovascular inflammation, underlie different stages of encephalopathy, from the early protracted stage to the late delayed stage. It is evident that ionizing radiation combined with other traumatic insults such as penetrating wound, burn, blast, systemic infection and chemotherapy, among others, can exacerbate the radiation sequelae (and vice versa) with increasing severity of neurogenic and microvascular patterns of radiation brain damage.
Collapse
Affiliation(s)
| | - Juliann G. Kiang
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
25
|
Bene BJ, Blakely WF, Burmeister DM, Cary L, Chhetri SJ, Davis CM, Ghosh SP, Holmes-Hampton GP, Iordanskiy S, Kalinich JF, Kiang JG, Kumar VP, Lowy RJ, Miller A, Naeem M, Schauer DA, Senchak L, Singh VK, Stewart AJ, Velazquez EM, Xiao M. Celebrating 60 Years of Accomplishments of the Armed Forces Radiobiology Research Institute1. Radiat Res 2021; 196:129-146. [PMID: 33979439 DOI: 10.1667/21-00064.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 04/20/2021] [Indexed: 11/03/2022]
Abstract
Chartered by the U.S. Congress in 1961, the Armed Forces Radiobiology Research Institute (AFRRI) is a Joint Department of Defense (DoD) entity with the mission of carrying out the Medical Radiological Defense Research Program in support of our military forces around the globe. In the last 60 years, the investigators at AFRRI have conducted exploratory and developmental research with broad application to the field of radiation sciences. As the only DoD facility dedicated to radiation research, AFRRI's Medical Radiobiology Advisory Team provides deployable medical and radiobiological subject matter expertise, advising commanders in the response to a U.S. nuclear weapon incident and other nuclear or radiological material incidents. AFRRI received the DoD Joint Meritorious Unit Award on February 17, 2004, for its exceptionally meritorious achievements from September 11, 2001 to June 20, 2003, in response to acts of terrorism and nuclear/radiological threats at home and abroad. In August 2009, the American Nuclear Society designated the institute a nuclear historic landmark as the U.S.'s primary source of medical nuclear and radiological research, preparedness and training. Since then, research has continued, and core areas of study include prevention, assessment and treatment of radiological injuries that may occur from exposure to a wide range of doses (low to high). AFRRI collaborates with other government entities, academic institutions, civilian laboratories and other countries to research the biological effects of ionizing radiation. Notable early research contributions were the establishment of dose limits for major acute radiation syndromes in primates, applicable to human exposures, followed by the subsequent evolution of radiobiology concepts, particularly the importance of immune collapse and combined injury. In this century, the program has been essential in the development and validation of prophylactic and therapeutic drugs, such as Amifostine, Neupogen®, Neulasta®, Nplate® and Leukine®, all of which are used to prevent and treat radiation injuries. Moreover, AFRRI has helped develop rapid, high-precision, biodosimetry tools ranging from novel assays to software decision support. New drug candidates and biological dose assessment technologies are currently being developed. Such efforts are supported by unique and unmatched radiation sources and generators that allow for comprehensive analyses across the various types and qualities of radiation. These include but are not limited to both 60Co facilities, a TRIGA® reactor providing variable mixed neutron and γ-ray fields, a clinical linear accelerator, and a small animal radiation research platform with low-energy photons. There are five major research areas at AFRRI that encompass the prevention, assessment and treatment of injuries resulting from the effects of ionizing radiation: 1. biodosimetry; 2. low-level and low-dose-rate radiation; 3. internal contamination and metal toxicity; 4. radiation combined injury; and 5. radiation medical countermeasures. These research areas are bolstered by an educational component to broadcast and increase awareness of the medical effects of ionizing radiation, in the mass-casualty scenario after a nuclear detonation or radiological accidents. This work provides a description of the military medical operations as well as the radiation facilities and capabilities present at AFRRI, followed by a review and discussion of each of the research areas.
Collapse
Affiliation(s)
| | | | | | - Lynnette Cary
- Scientific Research Department.,Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | | | - Catherine M Davis
- Scientific Research Department.,Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Sanchita P Ghosh
- Scientific Research Department.,Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Gregory P Holmes-Hampton
- Scientific Research Department.,Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Sergey Iordanskiy
- Scientific Research Department.,Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | | | - Juliann G Kiang
- Scientific Research Department.,Medicine.,Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | | | | | | | | | - David A Schauer
- Radiation Sciences Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | | | - Vijay K Singh
- Scientific Research Department.,Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | | | | | | |
Collapse
|
26
|
Kiang JG, Zhai M, Lin B, Smith JT, Anderson MN, Jiang S. Co-Therapy of Pegylated G-CSF and Ghrelin for Enhancing Survival After Exposure to Lethal Radiation. Front Pharmacol 2021; 12:628018. [PMID: 33603673 PMCID: PMC7884820 DOI: 10.3389/fphar.2021.628018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/07/2021] [Indexed: 12/17/2022] Open
Abstract
Exposure to ionizing radiation (radiation injury, RI) in nuclear-related episode is evident to be life-threatening. RI occurs at levels of organs, tissues, cytosols, or nucleus. Their mechanisms are still not fully understood. FDA approves pegylated granulocyte colony-stimulating factor (Neulasta™, Peg-G-CSF) for acute hematopoietic syndrome and has been shown to save lives after lethal RI. We aimed to test whether Ghrelin enhanced Peg-G-CSF's efficacy to save more lives after lethal RI. B6D2F1/J female mice were used for the study. They received 9.5 Gy (LD50/30 at 0.4 Gy/min) emitted from the 60Co-γ-photon radiation facility. Peg-G-CSF was injected subcutaneously at 1 mg/kg once on days 1, 8, and 15 after irradiation. Ghrelin contains 28 amino acid and is a hunger peptide that has been shown to stimulate food intake, promote intestinal epithelial cell proliferation, elevates immunity, inhibits brain hemorrhage, and increases stress-coping. Ghrelin was injected subcutaneously at 113 μg/kg once on days 1, 2, and 3 after irradiation. Survival, body weight, water consumption, hematology, spleen weight, splenocytes, bone marrow cells, and histology of bone marrow and ileum were performed. We observed that radiation resulted in 30-days survival by 30%. RI decreased their body weights and water consumption volumes. On the 30th day post-RI, platelets and WBCs such as basophils, eosinophils, monocytes, lymphocytes, neutrophils and leukocytes were still significantly decreased in surviving mice. Likewise, their RBC, hemoglobin, hematocrit, and splenocytes remained low; splenomegaly was found in these mice. Bone marrow in surviving RI animals maintained low cellularity with high counts of fat cells and low counts of megakaryocytes. Meanwhile, ileum histology displayed injury. However, mice co-treated with both drugs 24 h after RI resulted in 30-days survival by 45% above the vehicle group. Additionally, the body-weight loss was mitigated, the acute radiation syndrome was reduced. This co-therapy significantly increased neutrophils, eosinophils, leukocytes, and platelets in circulation, inhibited splenomegaly, and increased bone marrow cells. Histopathological analysis showed significant improvement on bone marrow cellularity and ileum morphology. In conclusion, the results provide a proof of concept and suggest that the co-therapy of Peg-G-CSF and Ghrelin is efficacious to ameliorate RI.
Collapse
Affiliation(s)
- Juliann G. Kiang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Min Zhai
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
| | - Bin Lin
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
| | - Joan T. Smith
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
| | - Marsha N. Anderson
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
| | - Suping Jiang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD, United States
| |
Collapse
|
27
|
Mesenchymal Stem Cells for Mitigating Radiotherapy Side Effects. Cells 2021; 10:cells10020294. [PMID: 33535574 PMCID: PMC7912747 DOI: 10.3390/cells10020294] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/19/2021] [Accepted: 01/29/2021] [Indexed: 12/14/2022] Open
Abstract
Radiation therapy for cancers also damages healthy cells and causes side effects. Depending on the dosage and exposure region, radiotherapy may induce severe and irreversible injuries to various tissues or organs, especially the skin, intestine, brain, lung, liver, and heart. Therefore, promising treatment strategies to mitigate radiation injury is in pressing need. Recently, stem cell-based therapy generates great attention in clinical care. Among these, mesenchymal stem cells are extensively applied because it is easy to access and capable of mesodermal differentiation, immunomodulation, and paracrine secretion. Here, we summarize the current attempts and discuss the future perspectives about mesenchymal stem cells (MSCs) for mitigating radiotherapy side effects.
Collapse
|
28
|
Liu Z, Bian M, Ma QQ, Zhang Z, Du HH, Wei CX. Design and Synthesis of New Benzo[d]oxazole-Based Derivatives and Their Neuroprotective Effects on β-Amyloid-Induced PC12 Cells. Molecules 2020; 25:E5391. [PMID: 33218007 PMCID: PMC7698601 DOI: 10.3390/molecules25225391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/15/2020] [Accepted: 11/16/2020] [Indexed: 12/14/2022] Open
Abstract
A series of novel synthetic substituted benzo[d]oxazole-based derivatives (5a-5v) exerted neuroprotective effects on β-amyloid (Aβ)-induced PC12 cells as a potential approach for the treatment of Alzheimer's disease (AD). In vitro studies show that most of the synthesized compounds were potent in reducing the neurotoxicity of Aβ25-35-induced PC12 cells at 5 μg/mL. We found that compound 5c was non-neurotoxic at 30 μg/mL and significantly increased the viability of Aβ25-35-induced PC12 cells at 1.25, 2.5 and 5 μg/mL. Western blot analysis showed that compound 5c promoted the phosphorylation of Akt and glycogen synthase kinase (GSK-3β) and decreased the expression of nuclear factor-κB (NF-κB) in Aβ25-35-induced PC12 cells. In addition, our findings demonstrated that compound 5c protected PC12 cells from Aβ25-35-induced apoptosis and reduced the hyperphosphorylation of tau protein, and decreased the expression of receptor for AGE (RAGE), β-site amyloid precursor protein (APP)-cleaving enzyme 1 (BACE1), inducible nitric oxide synthase (iNOS) and Bcl-2-associated X protein/B-cell lymphoma 2 (Bax/Bcl-2) via Akt/GSK-3β/NF-κB signaling pathway. In vivo studies suggest that compound 5c shows less toxicity than donepezil in the heart and nervous system of zebrafish.
Collapse
Affiliation(s)
- Zheng Liu
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao 028000, China; (Z.L.); (M.B.); (Q.-Q.M.)
| | - Ming Bian
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao 028000, China; (Z.L.); (M.B.); (Q.-Q.M.)
- Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao 028000, China
| | - Qian-Qian Ma
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao 028000, China; (Z.L.); (M.B.); (Q.-Q.M.)
- Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao 028000, China
| | - Zhuo Zhang
- College of Pharmaceutical Sciences, Yanbian University, Yanji 133022, China;
| | - Huan-Huan Du
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao 028000, China; (Z.L.); (M.B.); (Q.-Q.M.)
- Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao 028000, China
| | - Cheng-Xi Wei
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao 028000, China; (Z.L.); (M.B.); (Q.-Q.M.)
- Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao 028000, China
| |
Collapse
|
29
|
Mao L, Zhou Y, Chen L, Hu L, Liu S, Zheng W, Zhao J, Guo M, Chen C, He Z, Xu L. Identification of atypical mitogen-activated protein kinase MAPK4 as a novel regulator in acute lung injury. Cell Biosci 2020; 10:121. [PMID: 33088477 PMCID: PMC7570399 DOI: 10.1186/s13578-020-00484-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/10/2020] [Indexed: 12/15/2022] Open
Abstract
Background Acute lung injury (ALI) is a serious disease with highly morbidity and mortality that causes serious health problems worldwide. Atypical mitogen activated protein kinases (MAPKs) play critical roles in the development of tissues and have been proposed as promising therapeutic targets for various diseases. However, the potential role of atypical MAPKs in ALI remains elusive. In this study, we investigated the role of atypical MAPKs family member MAPK4 in ALI using LPS-induced murine ALI model. Results We found that MAPK4 deficiency mice exhibited prolonged survival time after LPS challenge, accompanied by alleviated pathology in lung tissues, decreased levels of pro-inflammatory cytokines and altered composition of immune cells in BALF. Furthermore, the transduction of related signaling pathways, including MK5, AKT, JNK, and p38 MAPK pathways, was reduced obviously in LPS-treated MAPK4−/− mice. Notably, the expression of MAPK4 was up-regulated in lung tissues of ALI model, which was not related with MAPK4 promoter methylation, but negatively orchestrated by transcriptional factors NFKB1 and NR3C1. Further studies have shown that the expression of MAPK4 was also increased in LPS-treated macrophages. Meanwhile, MAPK4 deficiency reduced the expression of related pro-inflammatory cytokines in macrophage in response to LPS treatment. Finally, MAPK4 knockdown using shRNA pre-treatment could ameliorate the pathology of lung tissues and prolong the survival time of mice after LPS challenge. Conclusions Collectively, these findings reveal an important biological function of atypical MAPK in mediating the pathology of ALI, indicating that MAPK4 might be a novel potential therapeutic target for ALI treatment.
Collapse
Affiliation(s)
- Ling Mao
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, 563003 Guizhou China.,Department of Immunology, Zunyi Medical University, Zunyi, 563003 Guizhou China
| | - Ya Zhou
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, 563003 Guizhou China.,Department of Medical Physics, Zunyi Medical University, Zunyi, 563003 Guizhou China
| | - Longqing Chen
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, 563003 Guizhou China.,Department of Immunology, Zunyi Medical University, Zunyi, 563003 Guizhou China
| | - Lin Hu
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, 563003 Guizhou China.,Department of Immunology, Zunyi Medical University, Zunyi, 563003 Guizhou China
| | - Shiming Liu
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, 563003 Guizhou China.,Department of Immunology, Zunyi Medical University, Zunyi, 563003 Guizhou China
| | - Wen Zheng
- Department of Laboratory Medicine, Qiannan Medical College for Nationalities, Guizhou, 558000 China
| | - Juanjuan Zhao
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, 563003 Guizhou China.,Department of Immunology, Zunyi Medical University, Zunyi, 563003 Guizhou China
| | - Mengmeng Guo
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, 563003 Guizhou China.,Department of Immunology, Zunyi Medical University, Zunyi, 563003 Guizhou China
| | - Chao Chen
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, 563003 Guizhou China.,Department of Immunology, Zunyi Medical University, Zunyi, 563003 Guizhou China
| | - Zhixu He
- Department of Paediatrics, Affiliated Hospital of Zunyi Medical University, Guizhou, 563000 China.,Key Laboratory of Adult Stem Cell Transformation Research, Chinese Academy of Medical Sciences, Guizhou, 563000 China
| | - Lin Xu
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, 563003 Guizhou China.,Department of Immunology, Zunyi Medical University, Zunyi, 563003 Guizhou China
| |
Collapse
|
30
|
Cannon G, Kiang JG. A review of the impact on the ecosystem after ionizing irradiation: wildlife population. Int J Radiat Biol 2020; 98:1054-1062. [PMID: 32663058 PMCID: PMC10139769 DOI: 10.1080/09553002.2020.1793021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE On 26 April 1986, reactor 4 at the Chernobyl power plant underwent a catastrophic failure leading to core explosions and open-air fires. On 11 March 2011, a combination of earthquake and tsunami led to a similar disaster at the Fukushima Daiichi power plant. In both cases, radioactive isotopes were released and contaminated the air, soil and water in a substantial area around the power plants. Humans were evacuated from the immediate regions but the wildlife stayed and continued to be affected by the ongoing high radiation exposure initially and later decayed amounts of fallout dusts with time. In this review, we will examine the significant effects of the increased radiation on vegetation, insects, fish, birds and mammals. CONCLUSIONS The initial intense radiation in these areas has gradually begun to decrease but still remains high. Adaptation to radiation is evident and the ecosystems have dynamically changed from the periods immediately after the accidents to the present day. Understanding the molecular mechanisms that allow the adaptation and recovery of wildlife to chronic radiation challenges would aid in future attempts at ecosystem remediation in the wake of such incidents.
Collapse
Affiliation(s)
- Georgetta Cannon
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Juliann G Kiang
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|