Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 836
1.
Gut Microbes ; 16(1): 2347722, 2024.
Article En | MEDLINE | ID: mdl-38706205

The intestine is prone to radiation damage in patients undergoing radiotherapy for pelvic tumors. However, there are currently no effective drugs available for the prevention or treatment of radiation-induced enteropathy (RIE). In this study, we aimed at investigating the impact of indole-3-carboxaldehyde (I3A) derived from the intestinal microbiota on RIE. Intestinal organoids were isolated and cultivated for screening radioprotective tryptophan metabolites. A RIE model was established using 13 Gy whole-abdominal irradiation in male C57BL/6J mice. After oral administration of I3A, its radioprotective ability was assessed through the observation of survival rates, clinical scores, and pathological analysis. Intestinal stem cell survival and changes in the intestinal barrier were observed through immunofluorescence and immunohistochemistry. Subsequently, the radioprotective mechanisms of I3A was investigated through 16S rRNA and transcriptome sequencing, respectively. Finally, human colon cancer cells and organoids were cultured to assess the influence of I3A on tumor radiotherapy. I3A exhibited the most potent radioprotective effect on intestinal organoids. Oral administration of I3A treatment significantly increased the survival rate in irradiated mice, improved clinical and histological scores, mitigated mucosal damage, enhanced the proliferation and differentiation of Lgr5+ intestinal stem cells, and maintained intestinal barrier integrity. Furthermore, I3A enhanced the abundance of probiotics, and activated the AhR/IL-10/Wnt signaling pathway to promote intestinal epithelial proliferation. As a crucial tryptophan metabolite, I3A promotes intestinal epithelial cell proliferation through the AhR/IL-10/Wnt signaling pathway and upregulates the abundance of probiotics to treat RIE. Microbiota-derived I3A demonstrates potential clinical application value for the treatment of RIE.


Gastrointestinal Microbiome , Indoles , Mice, Inbred C57BL , Probiotics , Receptors, Aryl Hydrocarbon , Wnt Signaling Pathway , Animals , Mice , Gastrointestinal Microbiome/drug effects , Male , Humans , Probiotics/administration & dosage , Probiotics/pharmacology , Receptors, Aryl Hydrocarbon/metabolism , Indoles/metabolism , Indoles/pharmacology , Radiation-Protective Agents/pharmacology , Organoids/metabolism , Radiation Injuries/metabolism , Radiation Injuries/prevention & control , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/radiation effects , Intestines/microbiology , Intestines/radiation effects , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics
2.
Microbiome ; 12(1): 89, 2024 May 14.
Article En | MEDLINE | ID: mdl-38745230

BACKGROUND: Non-toxic approaches to enhance radiotherapy outcomes are beneficial, particularly in ageing populations. Based on preclinical findings showing that high-fibre diets sensitised bladder tumours to irradiation by modifying the gut microbiota, along with clinical evidence of prebiotics enhancing anti-cancer immunity, we hypothesised that dietary fibre and its gut microbiota modification can radiosensitise tumours via secretion of metabolites and/or immunomodulation. We investigated the efficacy of high-fibre diets combined with irradiation in immunoproficient C57BL/6 mice bearing bladder cancer flank allografts. RESULT: Psyllium plus inulin significantly decreased tumour size and delayed tumour growth following irradiation compared to 0.2% cellulose and raised intratumoural CD8+ cells. Post-irradiation, tumour control positively correlated with Lachnospiraceae family abundance. Psyllium plus resistant starch radiosensitised the tumours, positively correlating with Bacteroides genus abundance and increased caecal isoferulic acid levels, associated with a favourable response in terms of tumour control. Psyllium plus inulin mitigated the acute radiation injury caused by 14 Gy. Psyllium plus inulin increased caecal acetate, butyrate and propionate levels, and psyllium alone and psyllium plus resistant starch increased acetate levels. Human gut microbiota profiles at the phylum level were generally more like mouse 0.2% cellulose profiles than high fibre profiles. CONCLUSION: These supplements may be useful in combination with radiotherapy in patients with pelvic malignancy. Video Abstract.


Dietary Fiber , Dietary Supplements , Gastrointestinal Microbiome , Inulin , Mice, Inbred C57BL , Psyllium , Urinary Bladder Neoplasms , Animals , Mice , Gastrointestinal Microbiome/drug effects , Inulin/administration & dosage , Urinary Bladder Neoplasms/radiotherapy , Urinary Bladder Neoplasms/pathology , Humans , Female , Radiation Injuries/prevention & control , Intestines/microbiology , Intestines/radiation effects , CD8-Positive T-Lymphocytes
3.
Int J Mol Sci ; 25(8)2024 Apr 20.
Article En | MEDLINE | ID: mdl-38674120

Hematopoietic acute radiation syndrome (H-ARS) involves injury to multiple organ systems following total body irradiation (TBI). Our laboratory demonstrated that captopril, an angiotensin-converting enzyme inhibitor, mitigates H-ARS in Göttingen minipigs, with improved survival and hematopoietic recovery, as well as the suppression of acute inflammation. However, the effects of captopril on the gastrointestinal (GI) system after TBI are not well known. We used a Göttingen minipig H-ARS model to investigate captopril's effects on the GI following TBI (60Co 1.79 or 1.80 Gy, 0.42-0.48 Gy/min), with endpoints at 6 or 35 days. The vehicle or captopril (0.96 mg/kg) was administered orally twice daily for 12 days, starting 4 h post-irradiation. Ilea were harvested for histological, protein, and RNA analyses. TBI increased congestion and mucosa erosion and hemorrhage, which were modulated by captopril. GPX-4 and SLC7A11 were downregulated post-irradiation, consistent with ferroptosis at 6 and 35 days post-irradiation in all groups. Interestingly, p21/waf1 increased at 6 days in vehicle-treated but not captopril-treated animals. An RT-qPCR analysis showed that radiation increased the gene expression of inflammatory cytokines IL1B, TNFA, CCL2, IL18, and CXCL8, and the inflammasome component NLRP3. Captopril suppressed radiation-induced IL1B and TNFA. Rectal microbiome analysis showed that 1 day of captopril treatment with radiation decreased overall diversity, with increased Proteobacteria phyla and Escherichia genera. By 6 days, captopril increased the relative abundance of Enterococcus, previously associated with improved H-ARS survival in mice. Our data suggest that captopril mitigates senescence, some inflammation, and microbiome alterations, but not ferroptosis markers in the intestine following TBI.


Acute Radiation Syndrome , Captopril , Disease Models, Animal , Ferroptosis , Gastrointestinal Microbiome , Inflammation , Swine, Miniature , Whole-Body Irradiation , Animals , Acute Radiation Syndrome/drug therapy , Swine , Inflammation/pathology , Captopril/pharmacology , Whole-Body Irradiation/adverse effects , Ferroptosis/drug effects , Gastrointestinal Microbiome/drug effects , Intestines/microbiology , Intestines/pathology , Intestines/drug effects , Intestines/radiation effects , Male , Angiotensin-Converting Enzyme Inhibitors/pharmacology
4.
Mol Nutr Food Res ; 68(8): e2300745, 2024 Apr.
Article En | MEDLINE | ID: mdl-38581304

SCOPE: Naringenin (NAR) possesses unique anti-inflammatory, antiapoptosis effects and various bioactivities; however, its role against radiation-induced intestinal injury (RIII) remains unclear. This study aims to investigate whether NAR has protective effects against radiation-induced intestinal injury and the underlying mechanisms. METHODS AND RESULTS: C57BL/6J mice are exposed to a single dose of 13 Gy X-ray total abdominal irradiation (TAI), then gavaged with NAR for 7 days. NAR treatment prolongs the survival rate, protects crypts and villi from damage, alleviates the level of radiation-induced inflammation, and mitigates intestinal barrier damage in the irradiated mice. Additionally, NAR reduces immune cell infiltration and intestinal epithelial cell apoptosis. NAR also shows radioprotective effects in human colon cancer cells (HCT116) and human intestinal epithelial cells (NCM460). It reduces cell damage by reducing intracellular calcium ion levels and reactive oxygen species (ROS) levels. NAR-mediated radioprotection is associated with the downregulation of transient receptor potential vanilloid 6 (TRPV6), and inhibition of apoptosis pathway. Notably, treatment with NAR fails to further increase the protective effects of the TRPV6 inhibitor 2-APB, indicating that TRPV6 inhibition is essential for NAR activity. CONCLUSION: NAR inhibits the apoptosis pathway by downregulating TRPV6 and reducing calcium ion level, thereby alleviating RIII. Therefore, NAR is a promising therapeutic drug for RIII.


Apoptosis , Flavanones , Mice, Inbred C57BL , Reactive Oxygen Species , TRPV Cation Channels , Animals , Flavanones/pharmacology , Humans , TRPV Cation Channels/metabolism , Apoptosis/drug effects , Reactive Oxygen Species/metabolism , Male , Mice , Radiation-Protective Agents/pharmacology , Intestinal Mucosa/drug effects , Intestinal Mucosa/radiation effects , Intestinal Mucosa/metabolism , HCT116 Cells , Calcium Channels/metabolism , Intestines/drug effects , Intestines/radiation effects , Calcium/metabolism , Radiation Injuries/drug therapy
5.
Sci Rep ; 14(1): 9906, 2024 04 30.
Article En | MEDLINE | ID: mdl-38689033

CUL4B, a crucial scaffolding protein in the largest E3 ubiquitin ligase complex CRL4B, is involved in a broad range of physiological and pathological processes. While previous research has shown that CUL4B participates in maintaining intestinal homeostasis and function, its involvement in facilitating intestinal recovery following ionizing radiation (IR) damage has not been fully elucidated. Here, we utilized in vivo and in vitro models to decipher the role of CUL4B in intestinal repair after IR-injury. Our findings demonstrated that prior to radiation exposure, CUL4B inhibited the ubiquitination modification of PSME3, which led to the accumulation of PSME3 and subsequent negative regulation of p53-mediated apoptosis. In contrast, after radiation, CUL4B dissociated from PSME3 and translocated into the nucleus at phosphorylated histones H2A (γH2AX) foci, thereby impeding DNA damage repair and augmenting p53-mediated apoptosis through inhibition of BRCA1 phosphorylation and RAD51. Our study elucidated the dynamic role of CUL4B in the repair of radiation-induced intestinal damage and uncovered novel molecular mechanisms underlying the repair process, suggesting a potential therapeutic strategy of intestinal damage after radiation therapy for cancers.


Apoptosis , Cullin Proteins , Intestines , Regeneration , Tumor Suppressor Protein p53 , Animals , Humans , Mice , Apoptosis/radiation effects , BRCA1 Protein/metabolism , BRCA1 Protein/genetics , Cullin Proteins/metabolism , Cullin Proteins/genetics , DNA Damage , DNA Repair , Histones/metabolism , Intestines/radiation effects , Intestines/pathology , Mice, Inbred C57BL , Phosphorylation/radiation effects , Rad51 Recombinase/metabolism , Radiation, Ionizing , Regeneration/radiation effects , Tumor Suppressor Protein p53/metabolism , Ubiquitination
6.
Radiat Res ; 199(3): 252-262, 2023 03 01.
Article En | MEDLINE | ID: mdl-36701761

Radiation-induced intestinal injury is one the most common adverse events of radiotherapy, which can severely affect quality of life. There are currently no effective preventive and therapeutic options for this disorder. Quercetin is a natural flavonoid found in common food species, with the characteristics of antioxidative, anti-inflammatory, and anti-cancerous activity. However, the role of quercetin on radiation-induced intestinal injury and the underlying mechanism remains poorly understood. In this study, we found quercetin treatment can improve the survival rate of mice after a single-dose (10 Gy) abdominal irradiation. Quercetin-pretreated mice significantly reduced radiation-induced DNA damage and intestinal epithelium cell apoptosis. In addition, quercetin also improved the proliferation activity of intestinal stem cells and promoted intestine epithelium repair after irradiation. Further studies demonstrated that quercetin treatment curtailed radiation-induced reactive oxygen species generation via regulating Nrf2 signaling in intestinal epithelium cells. Furthermore, treatment with Nrf2 inhibitor, could reverse the above effects. Altogether, quercetin can ameliorate radiation-induced intestine injury via regulating Nrf2 signaling, scavenging free radicals, and promoting intestinal epithelium repair.


Antioxidants , Radiation Injuries , Mice , Animals , Antioxidants/pharmacology , Quercetin/pharmacology , Quercetin/therapeutic use , NF-E2-Related Factor 2/genetics , Quality of Life , Intestines/radiation effects , Radiation Injuries/drug therapy , Radiation Injuries/prevention & control , Intestinal Mucosa , Regeneration
7.
Int J Mol Sci ; 23(9)2022 Apr 22.
Article En | MEDLINE | ID: mdl-35563033

The gastrointestinal (GI) system is highly susceptible to irradiation. Currently, there is no Food and Drug Administration (FDA)-approved medical countermeasures for GI radiation injury. The vitamin E analog gamma-tocotrienol (GT3) is a promising radioprotector in mice and nonhuman primates (NHP). We evaluated GT3-mediated GI recovery in total-body irradiated (TBI) NHPs. Sixteen rhesus macaques were divided into two groups; eight received vehicle and eight GT3 24 h prior to 12 Gy TBI. Proximal jejunum was assessed for structural injuries and crypt survival on day 4 and 7. Apoptotic cell death and crypt cell proliferation were assessed with TUNEL and Ki-67 immunostaining. Irradiation induced significant shortening of the villi and reduced mucosal surface area. GT3 induced an increase in crypt depth at day 7, suggesting that more stem cells survived and proliferated after irradiation. GT3 did not influence crypt survival after irradiation. GT3 treatment caused a significant decline in TUNEL-positive cells at both day 4 (p < 0.03) and 7 (p < 0.0003). Importantly, GT3 induced a significant increase in Ki-67-positive cells at day 7 (p < 0.05). These data suggest that GT3 has radioprotective function in intestinal epithelial and crypt cells. GT3 should be further explored as a prophylactic medical countermeasure for radiation-induced GI injury.


Acute Radiation Syndrome , Chromans , Radiation-Protective Agents , Vitamin E , Acute Radiation Syndrome/drug therapy , Acute Radiation Syndrome/prevention & control , Animals , Chromans/therapeutic use , Disease Models, Animal , Intestines/pathology , Intestines/radiation effects , Ki-67 Antigen , Macaca mulatta , Radiation-Protective Agents/pharmacology , Radiation-Protective Agents/therapeutic use , Vitamin E/analogs & derivatives , Vitamin E/therapeutic use
8.
Biochem Biophys Res Commun ; 595: 7-13, 2022 03 05.
Article En | MEDLINE | ID: mdl-35091109

The intestinal tract is an essential component of the body's immune system, and is extremely sensitive to exposure of ionizing radiation. While ionizing radiation can effectively induce multiple forms of cell death, whether it can also promote ferroptosis in intestinal cells and the possible interrelationship between ferroptosis and intestinal immune function has not been reported so far. Here, we found that radiation-induced major ultrastructural changes in mitochondria of small intestinal epithelial cells and the changes induced in iron content and MDA levels in the small intestine were consistent with that observed during cellular ferroptosis, thus suggesting occurrence of ferroptosis in radiation-induced intestinal damage. Moreover, radiation caused a substantial increase in the expression of ferroptosis-related factors such as LPCAT3 and ALOX15 mRNA, augmented the levels of immune-related factors INF-γ and TGF-ß mRNA, and decreased the levels of IL-17 mRNA thereby indicating that ionizing radiation induced ferroptosis and impairment of intestinal immune function. Liproxstatin-1 is a ferroptosis inhibitor that was found to ameliorate radiation-induced ferroptosis and promote the recovery from immune imbalances. These findings supported the role of ferroptosis in radiation-induced intestinal immune injury and provide novel strategies for protection against radiation injury through regulation of the ferroptosis pathway.


Ferroptosis/physiology , Intestines/pathology , Quinoxalines/pharmacology , Radiation Injuries, Experimental/prevention & control , Radiation, Ionizing , Spiro Compounds/pharmacology , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , 1-Acylglycerophosphocholine O-Acyltransferase/metabolism , Animals , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/metabolism , Ferroptosis/drug effects , Ferroptosis/radiation effects , Gene Expression/drug effects , Gene Expression/radiation effects , Glutathione/metabolism , Intestine, Small/drug effects , Intestine, Small/metabolism , Intestine, Small/radiation effects , Intestines/drug effects , Intestines/radiation effects , Male , Malondialdehyde/metabolism , Mice, Inbred BALB C , Microscopy, Electron, Transmission , Mitochondria/drug effects , Mitochondria/radiation effects , Mitochondria/ultrastructure , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/physiopathology , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/metabolism
9.
Biomed Pharmacother ; 146: 112350, 2022 Feb.
Article En | MEDLINE | ID: mdl-34952740

This study explored the radioprotective effects and possible underlying mechanisms of KR-31831 against radiation-induced injury in a mouse model. KR-31831 (30 and 50 mg/kg) was administered to mice 24 h and 30 min before exposure to a single lethal or sublethal dose of whole-body irradiation (WBI) (7 or 4 Gy, respectively). These animals were then evaluated for changes in mortality, various hematological and biochemical parameters, and histological features in response to these treatments. In addition, RNA sequencing was used to profile the radiation-induced transcriptomic response in the bone marrow cells. The results showed that KR-31831 dose-dependently prolonged the 30-day survival period and prevented damage to radiation-sensitive organs, such as the intestine and testis, in response to WBI. Damage to the hematopoietic system was also notably improved in the KR-31831-treated mice, as evidenced by an increase in bone marrow and peripheral blood cells, as well as recovery of the histopathological characteristics of the bone marrow. These protective effects were achieved, at least in part, via the suppression of radiation-induced increases in apoptotic cell death and erythropoietin levels in the plasma. Furthermore, the gene expression profiles of the bone marrow cells of the WBI-treated mice suggested that KR-31831 upregulates the expression of the genes involved in regulating apoptosis and modulating the immune response, both of which are required for protecting the bone marrow. These results suggest the potential therapeutic efficacy of KR-31831 for protection against radiation-induced injury.


Benzopyrans/therapeutic use , Imidazoles/therapeutic use , Radiation Injuries/drug therapy , Radiation-Protective Agents/therapeutic use , Whole-Body Irradiation/adverse effects , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/radiation effects , Intestines/drug effects , Intestines/radiation effects , Male , Mice, Inbred C57BL , Radiation Injuries/genetics , Testis/drug effects , Testis/radiation effects , Transcriptome/drug effects
10.
Nutrients ; 13(9)2021 Aug 24.
Article En | MEDLINE | ID: mdl-34578802

It is urgent to seek new potential targets for the prevention or relief of gastrointestinal syndrome in clinical radiation therapy for cancers. Vitamin D, mediated through the vitamin D receptor (VDR), has been identified as a protective nutrient against ionizing radiation (IR)-induced damage. This study investigated whether VDR could inhibit IR-induced intestinal injury and explored underlying mechanism. We first found that vitamin D induced VDR expression and inhibited IR-induced DNA damage and apoptosis in vitro. VDR was highly expressed in intestinal crypts and was critical for crypt stem/progenitor cell proliferation under physiological conditions. Next, VDR-deficient mice exposed to IR significantly increased DNA damage and crypt stem/progenitor cell apoptosis, leading to impaired intestinal regeneration as well as shorter survival time. Furthermore, VDR deficiency activated the Pmaip1-mediated apoptotic pathway of intestinal crypt stem/progenitor cells in IR-treated mice, whereas inhibition of Pmaip1 expression by siRNA transfection protected against IR-induced cell apoptosis. Therefore, VDR protects against IR-induced intestinal injury through inhibition of crypt stem/progenitor cell apoptosis via the Pmaip1-mediated pathway. Our results reveal the importance of VDR level in clinical radiation therapy, and targeting VDR may be a useful strategy for treatment of gastrointestinal syndrome.


Apoptosis/drug effects , Intestines/radiation effects , Radiation Injuries, Experimental/prevention & control , Receptors, Calcitriol/metabolism , Stem Cells/metabolism , Vitamin D/pharmacology , Animals , Cell Line , Cell Proliferation/drug effects , DNA Damage/drug effects , Intestinal Mucosa/metabolism , Intestines/drug effects , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-bcl-2/metabolism , Radiation Injuries, Experimental/pathology , Radiation-Protective Agents/pharmacology , Rats
12.
J BUON ; 26(2): 626-633, 2021.
Article En | MEDLINE | ID: mdl-34077015

PURPOSE: Despite the therapeutic effects of radiotherapy on tumor cells, it has potential severe adverse effects on the surrounding normal tissues. Acute or chronic intestinal adverse effects that are likely to occur in patients undergoing radiotherapy for pelvic and abdominal cancers lead to increased morbidity, significant impairment of the quality of life, and economic losses. Various biological, chemical and pharmacological agents are being tested to protect from and to treat radiation enteritis. This experimental study aimed to investigate the protective effects of melatonin against radiation-induced intestinal injury when administered before radiation exposure in rats. METHODS: In the present study, villus height and the number of villi in the ileum and jejunum of rats receiving two different doses of intraperitoneal melatonin (5 and 10 mg/kg) prior to a single fraction of radiation given at a dose of 8 Gy to the abdominal region, was evaluated by histopathological examination 3 and 7 days after radiation exposure. RESULTS: At a dose of 5 mg/kg, melatonin was found to be effective in preventing radiation-induced injury to villus height in the jejunum and the number of villi in the ileum and jejunum, and at a dose of 10 mg/kg it was also effective in preventing radiation-induced injury to villus height in the ileum. CONCLUSIONS: Melatonin is effective for the prevention of radiation-induced intestinal injury. This outcome can be considered an evidence to test melatonin in clinical trials.


Intestines/radiation effects , Melatonin/therapeutic use , Animals , Female , Humans , Male , Melatonin/pharmacology , Radiation Injuries/pathology , Rats , Rats, Sprague-Dawley
13.
Radiat Res ; 196(2): 204-212, 2021 08 01.
Article En | MEDLINE | ID: mdl-34043805

In the event of a radiological attack or accident, it is more likely that the absorbed radiation dose will be heterogeneous, rather than uniformly distributed throughout the body. This type of uneven dose distribution is known as partial-body irradiation (PBI). Partial exposure of the vital organs, specifically the highly radiosensitive intestines, may cause death, if the injury is significant and the post-exposure recovery is considerably compromised. Here we investigated the recovery rate and extent of recovery from PBI-induced intestinal damage in large animals. Rhesus macaques (Macaca mulatta) were randomly divided into four groups: sham-irradiated (0 Gy), 8 Gy PBI, 11 Gy PBI and 14 Gy PBI. A single dose of ionizing radiation was delivered in the abdominal region using a uniform bilateral anteroposterior and posteroanterior technique. Irradiated animals were scheduled for euthanasia on days 10, 28 or 60 postirradiation, and sham-irradiated animals on day 60. Intestinal structural injuries were assessed via crypt depth, villus height, and mucosal surface length in the four different intestinal regions (duodenum, proximal jejunum, distal jejunum and ileum) using H&E staining. Higher radiation doses corresponded with more injury at 10 days post-PBI and a faster recovery rate. However, at 60 days post-PBI, damage was still evident in all regions of the intestine. The proximal and distal ends (duodenum and ileum, respectively) sustained less damage and recovered more fully than the jejunum.


Duodenum/radiation effects , Ileum/radiation effects , Intestine, Small/radiation effects , Jejunum/radiation effects , Animals , Duodenum/physiopathology , Humans , Ileum/physiopathology , Intestinal Mucosa/physiopathology , Intestinal Mucosa/radiation effects , Intestine, Small/physiopathology , Intestines/physiopathology , Intestines/radiation effects , Jejunum/physiopathology , Macaca mulatta/physiology , Primates/physiology , Radiation Dosage , Radiation, Ionizing , Whole-Body Irradiation
14.
Life Sci ; 278: 119546, 2021 Aug 01.
Article En | MEDLINE | ID: mdl-33915129

AIMS: Intestinal injury is a clinical problem related to radiotherapy or accidental exposure to ionizing radiation. This study aimed to investigate the protective effect of p-coumaric acid (CA) against radiation induced intestinal injury. MAIN METHODS: The present study orally administered CA to C57BL/6 male mice at 30 min before total body irradiation and continued for 3 days post irradiation. Then, the mice were sacrificed at day 3.5 or 14 after irradiation, respectively. The blood was collected to analyze the inflammatory cytokines. The antioxidant indexes of jejunum tissues were determined. Hematoxylin and eosin staining and apoptosis analysis was studied to investigate the pathological changes of the jejunum tissues. In addition, quantitative real-time polymerase chain reaction (qRT-PCR) and western blot were carried out to determine the changes in mRNA and protein levels of jejunum tissues. KEY FINDINGS: Compared with the only irradiated group, treatment with CA improved intestinal morphology and apoptosis, increased the villus height and the ratio of villus height to crypt depth. It also reduced the oxidative stress and inflammatory response. The molecular mechanism analysis showed that CA significantly inhibited the pyroptosis genes (Caspase-1, NLRP3 and AIM2) mRNA expression and improved the intestinal barrier genes expression. SIGNIFICANCE: The results suggested that CA ameliorates ionizing radiation-induced intestinal injury by inhibition of oxidative stress, inflammatory response and pyroptosis.


Coumaric Acids/therapeutic use , Intestines/drug effects , Intestines/radiation effects , Oxidative Stress/drug effects , Radiation Injuries, Experimental/drug therapy , Radiation-Protective Agents/therapeutic use , Animals , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/radiation effects , Intestines/pathology , Male , Mice, Inbred C57BL , Pyroptosis/drug effects , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology , Radiation, Ionizing
15.
P R Health Sci J ; 40(1): 6-11, 2021 03.
Article En | MEDLINE | ID: mdl-33876912

Radiotherapy treatment-induced intestinal injury and gut microbial perturbation/dysbiosis have been implicated in the pathobiology of cancer-related fatigue. The objective of this brief review was to explore the available evidence of the relationship between intestinal injury and self-reported fatigue, especially among cancer patients. The scientific evidence-including our own-linking gut mucosal barrier dysfunction and gut microbial perturbation/dysbiosis induced by cancer treatment with worsening of cancer related fatigue (perhaps through the gut-brain axis) is limited but promising. Emerging data suggest that lifestyle interventions and the administration of specific probiotics may favorably modulate the gut microbiota and potentially mediate beneficial effects leading to improvements in fatigue.


Dysbiosis , Fatigue , Intestines/radiation effects , Neoplasms/radiotherapy , Radiation Injuries , Brain-Gut Axis , Humans , Intestines/injuries
16.
J Radiat Res ; 62(4): 618-625, 2021 Jul 10.
Article En | MEDLINE | ID: mdl-33912960

Metformin, a first-line oral drug for type II diabetes mellitus, not only reduces blood glucose levels, but also has many other biological effects. Recent studies have been conducted to determine the protective effect of metformin in irradiation injuries. However, the results are controversial and mainly focus on the time of metformin administration. In this study, we aimed to investigate the protective effect of metformin in BALB/c mice exposed to 6 Gy or 8 Gy of a 60Co source of γ-rays for total body irradiation (TBI). Survival outcomes were assessed following exposure to 8 Gy or 6 Gy TBI, and hematopoietic damage and intestinal injury were assessed after exposure to 6 Gy TBI. Metformin prolonged the survival of mice exposed to 8 Gy TBI and improved the survival rate of mice exposed to 6 Gy TBI only when administered before exposure to irradiation. Moreover, pretreatment with metformin reduced the frequency of micronuclei (MN) in the bone marrow of mice exposed to 6 Gy TBI. Pretreatment of metformin also protected the intestinal morphology of mice, reduced inflammatory response and decreased the number of apoptotic cells in intestine. In conclusion, we demonstrated that pretreatment with metformin could alleviate irradiation injury.


Metformin/pharmacology , Whole-Body Irradiation , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Erythrocytes/drug effects , Erythrocytes/metabolism , Hematopoiesis/drug effects , Hematopoiesis/radiation effects , Intestines/pathology , Intestines/radiation effects , Male , Mice, Inbred BALB C , Micronucleus, Germline/metabolism , Survival Analysis
17.
Int J Radiat Oncol Biol Phys ; 109(5): 1472-1482, 2021 04 01.
Article En | MEDLINE | ID: mdl-33714528

PURPOSE: Chemoradiation therapy is the standard of care in muscle-invasive bladder cancer (MIBC). Although agents such as gemcitabine can enhance tumor radiosensitivity, their side effects can limit patient eligibility and treatment efficacy. This study investigates ultrasound and microbubbles for targeting gemcitabine delivery to reduce normal-tissue toxicity in a murine orthotopic MIBC model. MATERIALS AND METHODS: CD1-nude mice were injected orthotopically with RT112 bladder tumor cells. Conventional chemoradiation involved injecting gemcitabine (10 mg/kg) before 6 Gy targeted irradiation of the bladder area using the Small Animal Radiation Research Platform (SARRP). Ultrasound-mediated gemcitabine delivery (10 mg/kg gemcitabine) involved either coadministration of microbubbles with gemcitabine or conjugating gemcitabine onto microbubbles followed by exposure to ultrasound (1.1 MHz center frequency, 1 MPa peak negative pressure, 1% duty cycle, and 0.5 Hz pulse repetition frequency) before SARRP irradiation. The effect of ultrasound and microbubbles alone was also tested. Tumor volumes were measured by 3D ultrasound imaging. Acute normal-tissue toxicity from 12 Gy to the lower bowel area was assessed using an intestinal crypt assay in mice culled 3.75 days posttreatment. RESULTS: A significant delay in tumor growth was observed with conventional chemoradiation therapy and both microbubble groups (P < .05 compared with the radiation-only group). Transient weight loss was seen in the microbubble groups, which resolved within 10 days posttreatment. A positive correlation was found between weight loss on day 3 posttreatment and tumor growth delay (P < .05; R2 = 0.76). In contrast with conventional chemoradiation therapy, ultrasound-mediated drug delivery methods did not exacerbate the acute intestinal toxicity using the crypt assay. CONCLUSIONS: Ultrasound and microbubbles offer a promising new approach for improving chemoradiation therapy for muscle-invasive bladder cancer, maintaining a delay in tumor growth but with reduced acute intestinal toxicity compared with conventional chemoradiation therapy.


Antimetabolites, Antineoplastic/administration & dosage , Chemoradiotherapy/adverse effects , Deoxycytidine/analogs & derivatives , Organs at Risk/radiation effects , Radiation Injuries/prevention & control , Urinary Bladder Neoplasms/therapy , Animals , Antimetabolites, Antineoplastic/adverse effects , Biotinylation , Cell Line, Tumor , Chemoradiotherapy/methods , Contrast Media/administration & dosage , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Deoxycytidine/chemical synthesis , Female , Humans , Intestines/radiation effects , Mice , Mice, Nude , Microbubbles , Neoplasm Invasiveness , Tumor Burden , Ultrasonography , Urinary Bladder/pathology , Urinary Bladder Neoplasms/diagnostic imaging , Urinary Bladder Neoplasms/pathology , Gemcitabine
18.
Int J Radiat Oncol Biol Phys ; 110(5): 1317-1324, 2021 08 01.
Article En | MEDLINE | ID: mdl-33771702

PURPOSE: Cervical cancer is a global health problem. Despite the growth of prevention programs, there is an important need to improve the effectiveness of treatment for patients with invasive, locally advanced disease. In this study we examined (1) the efficacy of radiation therapy (RT) with cisplatin (RTCT) and an orally administered CXCR4 inhibitor suitable for clinical use, X4-136; (2) biomarkers of response to RTCT and X4-136; and (3) intestinal toxicity from RTCT and X4-136. METHODS AND MATERIALS: Orthotopic cervical cancer xenografts derived from our patients were treated with RT (30 Gy; 2 Gy/d) and cisplatin (4 mg/kg/wk intraperitoneally) with or without concurrent X4-136 (100 mg/kg/d orally) for 3 weeks. Mice were euthanized immediately after treatment for biomarker assessment or followed to evaluate primary tumor growth delay and metastases. In separate experiments, acute and late intestinal injury were assessed histologically. RESULTS: RTCT alone increased CXCL12/CXCR4 signaling, intratumoral accumulation of myeloid cells, and PD-L1 expression. The addition of X4-136 during RTCT abrogated these effects, improved primary tumor response, and reduced metastases. Furthermore, X4-136 increased the proportion of surviving intestinal crypt cells after irradiation, in keeping with a reduction in acute RT toxicity, and reduced late histologic changes of late RT toxicity. CONCLUSIONS: The combination of RTCT and the CXCR4 inhibitor X4-136 improves cervical cancer primary tumor control and reduces lymph node metastases, while also reducing normal tissue injury associated with adverse intestinal effects. Few if any pharmacologic strategies have expanded the therapeutic window with RT, suggesting that this combination warrants testing in clinical trials. These benefits might apply to other tumors where RTCT plays a curative role.


Chemoradiotherapy/methods , Receptors, CXCR4/antagonists & inhibitors , Uterine Cervical Neoplasms/therapy , Animals , B7-H1 Antigen/metabolism , Cell Survival/drug effects , Cell Survival/radiation effects , Chemokine CXCL12/metabolism , Chemoradiotherapy/adverse effects , Cisplatin/therapeutic use , Female , Humans , Intestines/cytology , Intestines/drug effects , Intestines/radiation effects , Leukocyte Count , Lymphatic Metastasis , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Radiation Injuries, Experimental/prevention & control , Radiation-Sensitizing Agents/therapeutic use , Receptors, CXCR4/metabolism , Signal Transduction/drug effects , Signal Transduction/radiation effects , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
19.
Anticancer Res ; 41(3): 1365-1375, 2021 Mar.
Article En | MEDLINE | ID: mdl-33788728

BACKGROUND/AIM: Progress in cancer treatment and diagnosis has made second cancer after medical radiation exposure a particular concern among childhood cancer survivors. Calorie restriction (CR) is a broadly effective cancer prevention strategy, although its effects on radiation-induced intestinal tumours are unclear. Here we examined the cancer-preventative efficacy of a CR diet at different starting ages on radiation induction of intestinal tumours in mice. MATERIALS AND METHODS: Male C3B6F1 ApcMin/+ mice were irradiated with 0 or 2 Gy of X-rays at 2 weeks of age. After an interval of 2, 8 or 18 weeks, mice were fed with a non-CR (95 kcal/week/mouse) or CR (65 kcal/week/mouse) diet. Intestinal tumours were evaluated for number, size distribution and malignancy. RESULTS: CR suppressed the size and progression of both spontaneous and radiation-induced intestinal tumours depending on age at starting of CR. CR diets were effective even administered to adult mice. CONCLUSION: CR was effective for suppression of tumour progression, which was accelerated by radiation exposure. Use of CR might be a useful cancer-prevention strategy for radiation-induced tumours of the intestinal tract.


Caloric Restriction/methods , Diet , Intestinal Neoplasms/diagnosis , Neoplasms, Radiation-Induced/diagnosis , X-Rays , Age Factors , Animals , Disease Progression , Genes, APC , Intestinal Neoplasms/genetics , Intestines/pathology , Intestines/radiation effects , Male , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Radiation-Induced/genetics , Time Factors
20.
J Cell Mol Med ; 25(8): 3785-3792, 2021 04.
Article En | MEDLINE | ID: mdl-33609010

Severe ionizing radiation causes the acute lethal damage of haematopoietic system and gastrointestinal tract. Here, we found CL429, the novel chimeric TLR2/NOD2 agonist, exhibited significant radioprotective effects in mice. CL429 increased mice survival, protected mice against the lethal damage of haematopoietic system and gastrointestinal tract. CL429 was more effective than equivalent amounts of monospecific (TLR2 or NOD2) and combination (TLR2 + NOD2) of molecules in preventing radiation-induced death. The radioprotection of CL429 was mainly mediated by activating TLR2 and partially activating NOD2. CL429-induced radioprotection was largely dependent on the activation of TLR2-MyD88-NF-κB signalling pathway. In conclusion, the data suggested that the co-activation of TLR2 and NOD2 could induce significant synergistic radioprotective effects and CL429 might be a potential high-efficiency selective agent.


Acetylmuramyl-Alanyl-Isoglutamine/analogs & derivatives , Acute Radiation Syndrome/prevention & control , Hematopoietic System/drug effects , Intestines/drug effects , Nod2 Signaling Adaptor Protein/agonists , Radiation-Protective Agents/pharmacology , Toll-Like Receptor 2/agonists , Whole-Body Irradiation/adverse effects , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Acute Radiation Syndrome/etiology , Acute Radiation Syndrome/pathology , Animals , Hematopoietic System/radiation effects , Intestines/injuries , Intestines/radiation effects , Male , Mice , Mice, Inbred C57BL
...