Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.033
Filtrar
1.
BMC Cardiovasc Disord ; 24(1): 323, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38918713

RESUMEN

BACKGROUND: Radiotherapy is a primary local treatment for tumors, yet it may lead to complications such as radiation-induced heart disease (RIHD). Currently, there is no standardized approach for preventing RIHD. Dexmedetomidine (Dex) is reported to have cardio-protection effects, while its role in radiation-induced myocardial injury is unknown. In the current study, we aimed to evaluate the radioprotective effect of dexmedetomidine in X-ray radiation-treated mice. METHODS: 18 male mice were randomized into 3 groups: control, 16 Gy, and 16 Gy + Dex. The 16 Gy group received a single dose of 16 Gy X-ray radiation. The 16 Gy + Dex group was pretreated with dexmedetomidine (30 µg/kg, intraperitoneal injection) 30 min before X-ray radiation. The control group was treated with saline and did not receive X-ray radiation. Myocardial tissues were collected 16 weeks after X-ray radiation. Hematoxylin-eosin staining was performed for histopathological examination. Terminal deoxynucleotidyl transferase dUTP nick-end labeling staining was performed to assess the state of apoptotic cells. Immunohistochemistry staining was performed to examine the expression of CD34 molecule and von Willebrand factor. Besides, western blot assay was employed for the detection of apoptosis-related proteins (BCL2 apoptosis regulator and BCL2-associated X) as well as autophagy-related proteins (microtubule-associated protein 1 light chain 3, beclin 1, and sequestosome 1). RESULTS: The findings demonstrated that 16 Gy X-ray radiation resulted in significant changes in myocardial tissues, increased myocardial apoptosis, and activated autophagy. Pretreatment with dexmedetomidine significantly protects mice against 16 Gy X-ray radiation-induced myocardial injury by inhibiting apoptosis and autophagy. CONCLUSION: In summary, our study confirmed the radioprotective effect of dexmedetomidine in mitigating cardiomyocyte apoptosis and autophagy induced by 16 Gy X-ray radiation.


Asunto(s)
Apoptosis , Autofagia , Dexmedetomidina , Miocitos Cardíacos , Traumatismos Experimentales por Radiación , Animales , Autofagia/efectos de los fármacos , Autofagia/efectos de la radiación , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Miocitos Cardíacos/efectos de la radiación , Miocitos Cardíacos/metabolismo , Apoptosis/efectos de los fármacos , Masculino , Dexmedetomidina/farmacología , Traumatismos Experimentales por Radiación/prevención & control , Traumatismos Experimentales por Radiación/patología , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Protectores contra Radiación/farmacología , Modelos Animales de Enfermedad , Transducción de Señal/efectos de los fármacos , Ratones , Proteínas Relacionadas con la Autofagia/metabolismo , Ratones Endogámicos C57BL , Proteínas Reguladoras de la Apoptosis/metabolismo
2.
Biochem Biophys Res Commun ; 724: 150226, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-38865815

RESUMEN

In patients with high-level radiation exposure, gastrointestinal injury is the main cause of death. Despite the severity of damage to the gastrointestinal tract, no specific therapeutic option is available. Tauroursodeoxycholic acid (TUDCA) is a conjugated form of ursodeoxycholic acid that suppresses endoplasmic reticulum (ER) stress and regulates various cell-signaling pathways. We investigated the effect of TUDCA premedication in alleviating intestinal damage and enhancing the survival of C57BL/6 mice administered a lethal dose (15Gy) of focal abdominal irradiation. TUDCA was administered to mice 1 h before radiation exposure, and reduced apoptosis of the jejunal crypts 12 h after irradiation. At later timepoint (3.5 days), irradiated mice manifested intestinal morphological changes that were detected via histological examination. TUDCA decreased the inflammatory cytokine levels and attenuated the decrease in serum citrulline levels after radiation exposure. Although radiation induced ER stress, TUDCA pretreatment decreased ER stress in the irradiated intestinal cells. The effect of TUDCA indicates the possibility of radiation therapy for cancer in tumor cells. TUDCA did not affect cell proliferation and apoptosis in the intestinal epithelium. TUDCA decreased the invasive ability of the CT26 metastatic colon cancer cell line. Reduced invasion after TUDCA treatment was associated with decreased matrix metalloproteinase (MMP)-7 and MMP-13 expression, which play important roles in invasion and metastasis. This study shows a potential role of TUDCA in protecting against radiation-induced intestinal damage and inhibiting tumor cell migration without any radiation and radiation therapy effect.


Asunto(s)
Apoptosis , Estrés del Retículo Endoplásmico , Ratones Endogámicos C57BL , Protectores contra Radiación , Ácido Tauroquenodesoxicólico , Animales , Ácido Tauroquenodesoxicólico/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de la radiación , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Protectores contra Radiación/farmacología , Ratones , Masculino , Intestinos/efectos de la radiación , Intestinos/efectos de los fármacos , Intestinos/patología , Modelos Animales de Enfermedad , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/efectos de la radiación , Mucosa Intestinal/patología , Mucosa Intestinal/metabolismo , Traumatismos Experimentales por Radiación/prevención & control , Traumatismos Experimentales por Radiación/patología , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Traumatismos Experimentales por Radiación/metabolismo , Metaloproteinasa 13 de la Matriz/metabolismo , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación
3.
Sci Rep ; 14(1): 13315, 2024 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-38858439

RESUMEN

Exposure to high, marginally lethal doses or higher of ionizing radiation, either intentional or accidental, results in injury to various organs. Currently, there is only a limited number of safe and effective radiation countermeasures approved by US Food and Drug Administration for such injuries. These approved agents are effective for only the hematopoietic component of the acute radiation syndrome and must be administered only after the exposure event: currently, there is no FDA-approved agent that can be used prophylactically. The nutraceutical, gamma-tocotrienol (GT3) has been found to be a promising radioprotector of such exposure-related injuries, especially those of a hematopoietic nature, when tested in either rodents or nonhuman primates. We investigated the nature of injuries and the possible protective effects of GT3 within select organ systems/tissues caused by both non-lethal level (4.0 Gy), as well as potentially lethal level (5.8 Gy) of ionizing radiation, delivered as total-body or partial-body exposure. Results indicated that the most severe, dose-dependent injuries occurred within those organ systems with strong self-renewing capacities (e.g., the lymphohematopoietic and gastrointestinal systems), while in other tissues (e.g., liver, kidney, lung) endowed with less self-renewal, the pathologies noted tended to be less pronounced and less dependent on the level of exposure dose or on the applied exposure regimen. The prophylactic use of the test nutraceutical, GT3, appeared to limit the extent of irradiation-associated pathology within blood forming tissues and, to some extent, within the small intestine of the gastrointestinal tract. No distinct, global pattern of bodily protection was noted with the agent's use, although a hint of a possible radioprotective benefit was suggested not only by a lessening of apparent injury within select organ systems, but also by way of noting the lack of early onset of moribundity within select GT3-treated animals.


Asunto(s)
Suplementos Dietéticos , Protectores contra Radiación , Animales , Protectores contra Radiación/farmacología , Vitamina E/farmacología , Vitamina E/análogos & derivados , Síndrome de Radiación Aguda/prevención & control , Síndrome de Radiación Aguda/tratamiento farmacológico , Síndrome de Radiación Aguda/patología , Cromanos/farmacología , Masculino , Traumatismos Experimentales por Radiación/prevención & control , Traumatismos Experimentales por Radiación/patología , Macaca mulatta , Hígado/efectos de los fármacos , Hígado/efectos de la radiación , Hígado/patología
4.
Int J Radiat Oncol Biol Phys ; 119(4): 1234-1247, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38364948

RESUMEN

PURPOSE: Studies during the past 9 years suggest that delivering radiation at dose rates exceeding 40 Gy/s, known as "FLASH" radiation therapy, enhances the therapeutic index of radiation therapy (RT) by decreasing normal tissue damage while maintaining tumor response compared with conventional (or standard) RT. This study demonstrates the cardioprotective benefits of FLASH proton RT (F-PRT) compared with standard (conventional) proton RT (S-PRT), as evidenced by reduced acute and chronic cardiac toxicities. METHODS AND MATERIALS: Mice were imaged using cone beam computed tomography to precisely determine the heart's apex as the beam isocenter. Irradiation was conducted using a shoot-through technique with a 5-mm diameter circular collimator. Bulk RNA-sequencing was performed on nonirradiated samples, as well as apexes treated with F-PRT or S-PRT, at 2 weeks after a single 40 Gy dose. Inflammatory responses were assessed through multiplex cytokine/chemokine microbead assay and immunofluorescence analyses. Levels of perivascular fibrosis were quantified using Masson's Trichrome and Picrosirius red staining. Additionally, cardiac tissue functionality was evaluated by 2-dimensional echocardiograms at 8- and 30-weeks post-PRT. RESULTS: Radiation damage was specifically localized to the heart's apex. RNA profiling of cardiac tissues treated with PRT revealed that S-PRT uniquely upregulated pathways associated with DNA damage response, induction of tumor necrosis factor superfamily, and inflammatory response, and F-PRT primarily affected cytoplasmic translation, mitochondrion organization, and adenosine triphosphate synthesis. Notably, F-PRT led to a milder inflammatory response, accompanied by significantly attenuated changes in transforming growth factor ß1 and α smooth muscle actin levels. Critically, F-PRT decreased collagen deposition and better preserved cardiac functionality compared with S-PRT. CONCLUSIONS: This study demonstrated that F-PRT reduces the induction of an inflammatory environment with lower expression of inflammatory cytokines and profibrotic factors. Importantly, the results indicate that F-PRT better preserves cardiac functionality, as confirmed by echocardiography analysis, while also mitigating the development of long-term fibrosis.


Asunto(s)
Fibrosis , Cardiopatías , Inflamación , Terapia de Protones , Animales , Terapia de Protones/efectos adversos , Ratones , Inflamación/etiología , Inflamación/radioterapia , Cardiopatías/etiología , Cardiopatías/prevención & control , Cardiopatías/diagnóstico por imagen , Cardiopatías/radioterapia , Corazón/efectos de la radiación , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/prevención & control , Traumatismos Experimentales por Radiación/patología , Masculino , Traumatismos por Radiación/prevención & control
5.
Naunyn Schmiedebergs Arch Pharmacol ; 397(7): 5193-5205, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38252300

RESUMEN

Perturbations produced by ionizing radiation on intestinal tissue are considered one of highly drastic challenges in radiotherapy. Animals were randomized into five groups. The first group was allocated as control, and the second was subjected to whole body γ-irradiation (10 Gy). The third was administered HA NP (17.6 mg/kg/day; i.p.) and then irradiated. The fourth one received MitoQ (2 mg/kg/day; i.p.) and then irradiated. The last group received MitoQ/HA NP (2 mg/kg/day; i.p.) for 5 days prior to irradiation. Mice were sacrificed a week post-γ-irradiation for evaluation. MitoQ/HA NP ameliorated mitochondrial oxidative stress as indicated by rising (TAC) and glutathione peroxidase and decreasing malondialdehyde, showing its distinguished antioxidant yield. That impacted the attenuation of apoptosis, which was revealed by the restoration of the anti-apoptotic marker and lessening proapoptotic caspase-3. Inflammatory parameters dwindled via treatment with MitoQ/HA NP. Moreover, this new NP exerts its therapeutic action through a distinguished radioprotective pathway (Hmgb1/TLR-4.) Subsequently, these antioxidants and their nanoparticles conferred protection to intestinal tissue as manifested by histopathological examination. These findings would be associated with its eminent antioxidant potential through high mitochondria targeting, enhanced cellular uptake, and ROS scavenging. This research underlines MitoQ/HA NP as a new treatment for the modulation of intestinal damage caused by radiotherapy modalities.


Asunto(s)
Antioxidantes , Apoptosis , Rayos gamma , Ácido Hialurónico , Compuestos Organofosforados , Estrés Oxidativo , Protectores contra Radiación , Ubiquinona , Animales , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/efectos de la radiación , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Rayos gamma/efectos adversos , Ratones , Compuestos Organofosforados/farmacología , Masculino , Protectores contra Radiación/farmacología , Protectores contra Radiación/uso terapéutico , Antioxidantes/farmacología , Ubiquinona/análogos & derivados , Ubiquinona/farmacología , Ácido Hialurónico/farmacología , Traumatismos Experimentales por Radiación/prevención & control , Traumatismos Experimentales por Radiación/patología , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Traumatismos Experimentales por Radiación/metabolismo , Nanopartículas , Intestinos/efectos de los fármacos , Intestinos/efectos de la radiación , Intestinos/patología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/efectos de la radiación
6.
Biomed Pharmacother ; 163: 114808, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37146417

RESUMEN

Radiotherapy is a prevalent treatment modality for thoracic tumors; however, it can lead to radiation-induced lung injury (RILI), which currently lacks effective interventions. ACT001, a prodrug of micheliolide, has demonstrated promising clinical application potential, yet its impact on RILI requires further validation. This study aims to investigate the radioprotective effects of ACT001 on RILI and elucidate its underlying mechanism. Sprague-Dawley rats were utilized to induce RILI following 20 Gy X-ray chest irradiation, and lung tissue inflammation and fibrosis were assessed using hematoxylin and eosin (H&E) and Masson staining. Lung injury, inflammation, and oxidative stress markers were evaluated employing commercial kits. Pyroptosis-related differentially expressed genes (DEGs) were analyzed using a microarray dataset from the Gene Expression Omnibus (GEO) database, and their functions and hub genes were identified through protein-protein interaction networks. Pyroptosis-related genes were detected via RT-qPCR, western blotting, immunofluorescence, and immunohistochemistry. The results demonstrated that ACT001 ameliorated RILI, diminished pro-inflammatory cytokine release and fibrosis, and mitigated the activation of the NLRP3 inflammasome while inhibiting pyroptosis in lung tissue. In conclusion, our study reveals that ACT001 can suppress NLRP3 inflammasome-mediated pyroptosis and improve RILI, suggesting its potential as a novel protective agent for RILI.


Asunto(s)
Lesión Pulmonar , Traumatismos Experimentales por Radiación , Ratas , Animales , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/prevención & control , Lesión Pulmonar/patología , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ratas Sprague-Dawley , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Traumatismos Experimentales por Radiación/prevención & control , Rayos X , Inflamación
7.
J Ethnopharmacol ; 311: 116428, 2023 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-36997130

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Irradiation-induced intestinal injury (RIII) often occurs during radiotherapy in patients, which would result in abdominal pain, diarrhea, nausea, vomiting, and even death. Engelhardia roxburghiana Wall. leaves, a traditional Chinese herb, has unique anti-inflammatory, anti-tumor, antioxidant, and analgesic effects, is used to treat damp-heat diarrhea, hernia, and abdominal pain, and has the potential to protect against RIII. AIM OF THE STUDY: To explore the protective effects of the total flavonoids of Engelhardia roxburghiana Wall. leaves (TFERL) on RIII and provide some reference for the application of Engelhardia roxburghiana Wall. leaves in the field of radiation protection. MATERIALS AND METHODS: The effect of TFERL on the survival rate of mice was observed after a lethal radiation dose (7.2 Gy) by ionizing radiation (IR). To better observe the protective effects of the TFERL on RIII, a mice model of RIII induced by IR (13 Gy) was established. Small intestinal crypts, villi, intestinal stem cells (ISC) and the proliferation of ISC were observed by haematoxylin and eosin (H&E) and immunohistochemistry (IHC). Quantitative real-time PCR (qRT-PCR) was used to detect the expression of genes related to intestinal integrity. Superoxide dismutase (SOD), reduced glutathione (GSH), interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) in the serum of mice were assessed. In vitro, cell models of RIII induced by IR (2, 4, 6, 8 Gy) were established. Normal human intestinal epithelial cells HIEC-6 cells were treated with TFERL/Vehicle, and the radiation protective effect of TFERL on HIEC-6 cells was detected by clone formation assay. DNA damage was detected by comet assay and immunofluorescence assay. Reactive oxygen species (ROS), cell cycle and apoptosis rate were detected by flow cytometry. Oxidative stress, apoptosis and ferroptosis-related proteins were detected by western blot. Finally, the colony formation assay was used to detect the effect of TFERL on the radiosensitivity of colorectal cancer cells. RESULTS: TFERL treatment can increase the survival rate and time of the mice after a lethal radiation dose. In the mice model of RIII induced by IR, TFERL alleviated RIII by reducing intestinal crypt/villi structural damage, increasing the number and proliferation of ISC, and maintaining the integrity of the intestinal epithelium after total abdominal irradiation. Moreover, TFERL promoted the proliferation of irradiated HIEC-6 cells, and reduced radiation-induced apoptosis and DNA damage. Mechanism studies have found that TFERL promotes the expression of NRF2 and its downstream antioxidant proteins, and silencing NRF2 resulted in the loss of radioprotection by TFERL, suggesting that TFERL exerts radiation protection by activating the NRF2 pathway. Surprisingly, TFERL reduced the number of clones of colon cancer cells after irradiation, suggesting that TFERL can increase the radiosensitivity of colon cancer cells. CONCLUSION: Our data showed that TFERL inhibited oxidative stress, reduced DNA damage, reduced apoptosis and ferroptosis, and improved IR-induced RIII. This study may offer a fresh approach to using Chinese herbs for radioprotection.


Asunto(s)
Neoplasias del Colon , Traumatismos Experimentales por Radiación , Humanos , Animales , Ratones , Antioxidantes/farmacología , Factor 2 Relacionado con NF-E2 , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Traumatismos Experimentales por Radiación/prevención & control , Apoptosis , Diarrea , Dolor Abdominal
8.
Int J Radiat Biol ; 99(7): 1119-1129, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36794325

RESUMEN

PURPOSE: To test IPW-5371 for the mitigation of the delayed effects of acute radiation exposure (DEARE). Survivors of acute radiation exposure are at risk for developing delayed multi-organ toxicities; however, there are no FDA-approved medical countermeasures (MCM) to mitigate DEARE. METHODS: WAG/RijCmcr female rat model of partial-body irradiation (PBI), by shielding part of one hind leg, was used to test IPW-5371 (7 and 20 mg kg-1 d-1) for mitigation of lung and kidney DEARE when started 15 d after PBI. Rats were fed known amounts of IPW-5371 using a syringe, instead of delivery by daily oral gavage, sparing exacerbation of esophageal injury by radiation. The primary endpoint, all-cause morbidity was assessed over 215 d. Secondary endpoints: body weight, breathing rate and blood urea nitrogen were also assessed. RESULTS: IPW-5371 enhanced survival (primary endpoint) as well as attenuated secondary endpoints of lung and kidney injuries by radiation. CONCLUSION: To provide a window for dosimetry and triage, as well as avoid oral delivery during the acute radiation syndrome (ARS), the drug regimen was started at 15 d after 13.5 Gy PBI. The experimental design to test mitigation of DEARE was customized for translation in humans, using an animal model of radiation that was designed to simulate a radiologic attack or accident. The results support advanced development of IPW-5371 to mitigate lethal lung and kidney injuries after irradiation of multiple organs.


Asunto(s)
Síndrome de Radiación Aguda , Traumatismos Experimentales por Radiación , Humanos , Ratas , Femenino , Animales , Traumatismos Experimentales por Radiación/prevención & control , Médula Ósea/efectos de la radiación , Dosis de Radiación , Pulmón/efectos de la radiación
9.
Int J Radiat Biol ; 99(2): 259-269, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35583501

RESUMEN

PURPOSE: With the development of nuclear technology and radiotherapy, the risk of radiation injury has been increasing. Therefore, it is important to find an effective radiation-protective agent. In this study, we designed and synthesized a novel compound called compound 8, of which the radioprotective effect and mechanism were studied. MATERIALS AND METHODS: Before being exposed to ionizing radiation, mice were pretreated with compound 8. The 30-day mortality assay, hematoxylin-eosin staining, and immunohistochemistry staining assay were performed to evaluate the anti-radiation effect of the compound 8. TUNEL and immunofluorescence assays were conducted to study the anti-radiation mechanism of compound 8. RESULTS: Compared to the IR + vehicle group, the 30-day survival rate of mice treated with 25 mg/kg of compound 8 was significantly improved after 8 Gy total body irradiation. In the morphological study of the small intestine, we found that compound 8 could maintain crypt-villus structures in the irradiated mice. Further immunohistochemical staining displayed that compound 8 could improve the survival of Lgr5+ cells, ki67+ cells, and lysozyme+ cells. The results of TUNEL and immunofluorescence assays showed that compound 8 could decrease the expression of apoptosis-related caspase-8/-9, γ-H2AX, Bax, and p53. CONCLUSIONS: These results indicate that compound 8 exerts its effects by maintaining structure and function of small intestine. It also reduces DNA damage, promotes crypt proliferation and differentiation. Moreover, it may enhance the anti-apoptotic ability of small intestinal tissue by inhibiting the activation of p53 and blocking the caspase cascade reaction. Compound 8 can protect the intestinal tract from post-radiation damage, it is thus a new and effective protective agent of radiation.


Asunto(s)
Traumatismos Experimentales por Radiación , Protectores contra Radiación , Ratones , Animales , Proteína p53 Supresora de Tumor/metabolismo , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Traumatismos Experimentales por Radiación/prevención & control , Traumatismos Experimentales por Radiación/metabolismo , Intestino Delgado , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efectos de la radiación , Radiación Ionizante , Protectores contra Radiación/farmacología , Protectores contra Radiación/química , Apoptosis/efectos de la radiación , Ratones Endogámicos C57BL
10.
Front Immunol ; 13: 927213, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36110845

RESUMEN

Recently, Toll-like receptors (TLRs) have been extensively studied in radiation damage, but the inherent defects of high toxicity and low efficacy of most TLR ligands limit their further clinical transformation. CRX-527, as a TLR4 ligand, has rarely been reported to protect against radiation. We demonstrated that CRX-527 was safer than LPS at the same dose in vivo and had almost no toxic effect in vitro. Administration of CRX-527 improved the survival rate of total body irradiation (TBI) to 100% in wild-type mice but not in TLR4-/- mice. After TBI, hematopoietic system damage was significantly alleviated, and the recovery period was accelerated in CRX-527-treated mice. Moreover, CRX-527 induced differentiation of HSCs and the stimulation of CRX-527 significantly increased the proportion and number of LSK cells and promoted their differentiation into macrophages, activating immune defense. Furthermore, we proposed an immune defense role for hematopoietic differentiation in the protection against intestinal radiation damage, and confirmed that macrophages invaded the intestines through peripheral blood to protect them from radiation damage. Meanwhile, CRX-527 maintained intestinal function and homeostasis, promoted the regeneration of intestinal stem cells, and protected intestinal injury from lethal dose irradiation. Furthermore, After the use of mice, we found that CRX-527 had no significant protective effect on the hematopoietic and intestinal systems of irradiated TLR4-/- mice. in conclusion, CRX-527 induced differentiation of HSCs protecting the intestinal epithelium from radiation damage.


Asunto(s)
Células Madre Hematopoyéticas , Compuestos Organofosforados , Traumatismos Experimentales por Radiación , Receptor Toll-Like 4 , Animales , Apoptosis , Diferenciación Celular , Glucosamina/análogos & derivados , Glucosamina/farmacología , Células Madre Hematopoyéticas/citología , Mucosa Intestinal , Ligandos , Lipopolisacáridos/farmacología , Ratones , Compuestos Organofosforados/farmacología , Traumatismos Experimentales por Radiación/prevención & control , Receptor Toll-Like 4/genética
11.
Biochem Biophys Res Commun ; 595: 7-13, 2022 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-35091109

RESUMEN

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.


Asunto(s)
Ferroptosis/fisiología , Intestinos/patología , Quinoxalinas/farmacología , Traumatismos Experimentales por Radiación/prevención & control , Radiación Ionizante , Compuestos de Espiro/farmacología , 1-Acilglicerofosfocolina O-Aciltransferasa/genética , 1-Acilglicerofosfocolina O-Aciltransferasa/metabolismo , Animales , Araquidonato 12-Lipooxigenasa/genética , Araquidonato 12-Lipooxigenasa/metabolismo , Araquidonato 15-Lipooxigenasa/genética , Araquidonato 15-Lipooxigenasa/metabolismo , Ferroptosis/efectos de los fármacos , Ferroptosis/efectos de la radiación , Expresión Génica/efectos de los fármacos , Expresión Génica/efectos de la radiación , Glutatión/metabolismo , Intestino Delgado/efectos de los fármacos , Intestino Delgado/metabolismo , Intestino Delgado/efectos de la radiación , Intestinos/efectos de los fármacos , Intestinos/efectos de la radiación , Masculino , Malondialdehído/metabolismo , Ratones Endogámicos BALB C , Microscopía Electrónica de Transmisión , Mitocondrias/efectos de los fármacos , Mitocondrias/efectos de la radiación , Mitocondrias/ultraestructura , Traumatismos Experimentales por Radiación/patología , Traumatismos Experimentales por Radiación/fisiopatología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Superóxido Dismutasa/metabolismo
12.
Biomed Pharmacother ; 146: 112496, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34959117

RESUMEN

Intestinal injury is one of the major side effects that are induced by medical radiation exposure, and has limited effective therapies. In this study, we investigated the beneficial effects of sanguinarine (SAN) on intestinal injury induced by ionizing radiation (IR) both in vitro and in vivo. Mice were exposed to whole abdominal irradiation (WAI) to mimic clinical scenarios. SAN was injected intraperitoneally to mitigate IR-induced injury. Histological examination was performed to assess the tissue injuries of the spleen and small intestine. A small intestinal epithelial cell line-6 (IEC-6) was analyzed for its viability and apoptosis in vitro under different treatments. Inflammation-related pathways and serum inflammatory cytokines were detected via Western blot analysis and ELISA, respectively. High-throughput sequencing was used to characterize the gut microbiota profile. High-performance liquid chromatography was performed to assess short-chain fatty acid contents in the colon. In vitro, SAN pretreatment protected cell viability and reduced apoptosis in IEC-6 cells. In vivo, SAN pretreatment protected immune organs, alleviated intestinal injury, and promoted intestinal recovery. SAN also reduced the levels of inflammatory cytokines, suppressed high mobility group box 1 (HMGB1)/ Toll-like receptor 4 (TLR4) pathway activation, and modulated gut microbiota composition. Our findings demonstrate that the beneficial properties of SAN alleviated intestinal radiation injury. Thus, SAN represents a therapeutic option for protecting against IR-induced intestinal injury in preclinical settings.


Asunto(s)
Benzofenantridinas/farmacología , Intestino Delgado/efectos de los fármacos , Isoquinolinas/farmacología , Traumatismos Experimentales por Radiación/prevención & control , Bazo/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Citocinas/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Ácidos Grasos Volátiles , Microbioma Gastrointestinal/efectos de los fármacos , Proteína HMGB1/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Inyecciones Intraperitoneales , Intestino Delgado/patología , Intestino Delgado/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos C57BL , Traumatismos Experimentales por Radiación/patología , Radiación Ionizante , Transducción de Señal/efectos de los fármacos , Bazo/patología , Bazo/efectos de la radiación , Receptor Toll-Like 4/efectos de los fármacos
13.
Oxid Med Cell Longev ; 2021: 2231680, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34873428

RESUMEN

Radiation-induced oral mucositis is a major adverse event of radiotherapy. Severe oral mucositis may cause unwanted interruption in radiotherapy and reduce long-term survival in cancer patients receiving radiotherapy, but until now, there have been no effective options for preventing radiation-induced oral mucositis. Quercetin is a flavonoid that is widely found in food species and has anti-inflammatory, antioxidant, and anticancer activities. In this study, we investigated a new role of quercetin in preventing radiation-induced oral mucositis. Quercetin exerted preventive effects against radiation-induced oral mucositis induced by single-dose (25 Gy) ionizing radiation or fractionated ionizing radiation (8 Gy × 3) in C57BL/6 mice and maintained the proliferation ability of basal epithelial cells. Quercetin pretreatment alleviated reactive oxygen species generation, NF-κB pathway activation, and downstream proinflammatory cytokine production and reduced DNA double-strand breaks and cellular senescence induced by ionizing radiation. Quercetin also upregulated BMI-1 expression in oral epithelial cells and promoted ulcer repair. In addition, quercetin exerted similar radioprotective effects in irradiated primary cultured normal human keratinocytes, reduced reactive oxygen species generation and proinflammatory cytokine release, and promoted DNA double-strand break repair and wound healing by upregulating the expression of BMI-1, which is a polycomb group protein. Thus, quercetin can block multiple pathological processes of radiation-induced oral mucositis by targeting BMI-1 and may be a potential treatment option for preventing radiation-induced oral mucositis.


Asunto(s)
Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Quercetina/farmacología , Traumatismos Experimentales por Radiación/prevención & control , Estomatitis/prevención & control , Animales , Antioxidantes/farmacología , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Traumatismos Experimentales por Radiación/metabolismo , Distribución Aleatoria , Estomatitis/etiología , Estomatitis/metabolismo , Regulación hacia Arriba/efectos de los fármacos
14.
Nutrients ; 13(9)2021 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-34578802

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Intestinos/efectos de la radiación , Traumatismos Experimentales por Radiación/prevención & control , Receptores de Calcitriol/metabolismo , Células Madre/metabolismo , Vitamina D/farmacología , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Traumatismos Experimentales por Radiación/patología , Protectores contra Radiación/farmacología , Ratas
15.
Health Phys ; 121(4): 419-433, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34546222

RESUMEN

ABSTRACT: The goal of this study was to develop rat models of partial body irradiation with bone-marrow sparing (leg-out PBI) to test medical countermeasures (MCM) of both acute radiation syndrome (ARS) and delayed effects of acute radiation exposure (DEARE) under the FDA animal rule. The leg-out PBI models were developed in female and male WAG/RijCmcr rats at doses of 12.5-14.5 Gy. Rats received supportive care consisting of fluids and antibiotics. Gastrointestinal ARS (GI-ARS) was assessed by lethality to d 7 and diarrhea scoring to d 10. Differential blood counts were analyzed between d 1-42 for the natural history of hematopoietic ARS (H-ARS). Lethality and breathing intervals (BI) were measured between d 28-110 to assess delayed injury to the lung (L-DEARE). Kidney injury (K-DEARE) was evaluated by measuring elevation of blood urea nitrogen (BUN) between d 90-180. The LD50/30, including both lethality from GI-ARS and H-ARS, for female and male rats are 14.0 Gy and 13.5 Gy, respectively, while the LD50/7 for only GI-ARS are 14.3 Gy and 13.6 Gy, respectively. The all-cause mortalities, including ARS and L-DEARE, through 120 d (LD50/120) are 13.5 Gy and 12.9 Gy, respectively. Secondary end points confirmed occurrence of four distinct sequelae representing GI, hematopoietic, lung, and kidney toxicities after leg-out PBI. Adult rat models of leg-out PBI showed the acute and long-term sequelae of radiation damage that has been reported in human radiation exposure case studies. Sex-specific differences were observed in the DRR between females and males. These rat models are among the most useful for the development and approval of countermeasures for mitigation of radiation injuries under the FDA animal rule.


Asunto(s)
Síndrome de Radiación Aguda , Sistema Hematopoyético , Contramedidas Médicas , Exposición a la Radiación , Traumatismos Experimentales por Radiación , Síndrome de Radiación Aguda/tratamiento farmacológico , Síndrome de Radiación Aguda/etiología , Síndrome de Radiación Aguda/prevención & control , Animales , Médula Ósea/efectos de la radiación , Femenino , Masculino , Traumatismos Experimentales por Radiación/complicaciones , Traumatismos Experimentales por Radiación/prevención & control , Ratas
16.
Oxid Med Cell Longev ; 2021: 4714649, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34471464

RESUMEN

The risk of radiation damage has increased with the rapid development of nuclear technology and radiotherapy. Hence, research on radioprotective agents is of utmost importance. In the present study, a novel aminothiol compound 12, containing a linear alkylamino backbone and three terminal thiols, was synthesized. Owing to the appropriate capped groups in the chains, it has an improved permeability and oral bioavailability compared to other radioprotective agents. Oral administration of compound 12 improved the survival of mice that received lethal doses of γ-irradiation. Experimental results demonstrated that compound 12 not only mitigated total body irradiation-induced hematopoietic injury by increasing the frequencies of hematopoietic stem and progenitor cells but also prevented abdominal irradiation-induced intestinal injury by increasing the survival of Lgr5+ intestinal cells, lysozyme+ Paneth cells, and Ki67+ cells. In addition, compound 12 decreased oxidative stress by upregulating the expression of Nrf2 and NQO1 and downregulating the expression of NOX1. Further, compound 12 inhibited γ-irradiation-induced DNA damage and alleviated G2/M phase arrest. Moreover, compound 12 decreased the levels of p53 and Bax and increased the level of Bcl-2, demonstrating that it may suppress radiation-induced apoptosis via the p53 pathway. These results indicate that compound 12 has the possibility of preventing radiation injury and can be a potential radioprotector for clinical applications.


Asunto(s)
Traumatismos Experimentales por Radiación/prevención & control , Protectores contra Radiación/uso terapéutico , Compuestos de Sulfhidrilo/uso terapéutico , Animales , Humanos , Masculino , Ratones , Protectores contra Radiación/farmacología , Compuestos de Sulfhidrilo/farmacología
17.
Sci Rep ; 11(1): 18356, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34526618

RESUMEN

Glycyrrhizin (GL) is a direct inhibitor of HMGB1 which acts as an alarmin when excreted into the extracellular space. High-dose radiation in radiotherapy induces collateral damage to the normal tissue, which can be mitigated by GL inhibiting HMGB1. The purpose of this study was to assess changes in HMGB1 and pro-inflammatory cytokines and to evaluate the protective effect of GL after low-dose radiation exposure. BALB/c mice were irradiated with 0.1 Gy (n = 10) and 1 Gy (n = 10) with GL being administered to half of the mice (n = 5, respectively) before irradiation. Blood and spleen samples were harvested and assessed for oxidative stress, HMGB1, pro-inflammatory cytokines, and cell viability. HMGB1 and pro-inflammatory cytokines increased and cell viability decreased after irradiation in a dose-dependent manner. Oxidative stress also increased after irradiation, but did not differ between 0.1 Gy and 1 Gy. With the pretreatment of GL, oxidative stress, HMGB1, and all of the pro-inflammatory cytokines decreased while cell viability was preserved. Our findings indicate that even low-dose radiation can induce sterile inflammation by increasing serum HMGB1 and pro-inflammatory cytokines and that GL can ameliorate the sterile inflammatory process by inhibiting HMGB1 to preserve cell viability.


Asunto(s)
Antiinflamatorios/farmacología , Ácido Glicirrínico/farmacología , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Bazo/efectos de los fármacos , Animales , Antiinflamatorios/uso terapéutico , Supervivencia Celular , Células Cultivadas , Citocinas/sangre , Ácido Glicirrínico/uso terapéutico , Proteína HMGB1/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos BALB C , Estrés Oxidativo , Traumatismos Experimentales por Radiación/prevención & control , Radiación Ionizante , Bazo/efectos de la radiación
18.
Biomed Pharmacother ; 142: 112064, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34426255

RESUMEN

In the work, the effect of black tea on oxidative stress induced in the aorta by irradiation was studied. The efficiency of black and green tea types was compared, and the effect of the main green tea components (-)-epigallocatechin galate (EGCG) and (-)-epigallocatechin (EGC) on the aorta was studied. The activity of ACE in rat aorta segments was determined by measuring the hydrolysis of hippuryl-L-histidyl-L-leucine, and the production of ROS was estimated from the oxidation of dichlorodihydrofluorescein. Black tea prevented the radiation-induced activation of the ACE and suppressed increased ROS production in the aorta of irradiated rats. The IC50 value for the suppression of the irradiation-induced increase in ACE activity is 1 ml of black tea brewed at a rate of 0.17 g/100 ml. Black tea is 12 times more effective than green tea. The administration of both catechin derivatives from green tea to rats leads to an increase in the activity of ACE and the formation of ROS in the aorta. The dose that provided half maximum activation of ACE (EC50) on intraperitoneal (i. p.) injection of galloylated catechins was found to be the same, 0.06-0.07 µg/kg of body weight. Upon intragastric gavage of EGCG, the EC50 value was by one order of magnitude higher, 0.8 µg/kg. Black tea was more effective than green tea in prevention a radiation-induced increase of ACE activity and oxidative stress in the aorta. This difference was explained by a low content of galloylated catechins in black tea.


Asunto(s)
Estrés Oxidativo/efectos de la radiación , Traumatismos Experimentales por Radiación/prevención & control , Protectores contra Radiación/farmacología , Té/química , Animales , Aorta/patología , Aorta/efectos de la radiación , Catequina/análogos & derivados , Catequina/aislamiento & purificación , Catequina/farmacología , Concentración 50 Inhibidora , Masculino , Protectores contra Radiación/aislamiento & purificación , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo
19.
Exp Eye Res ; 211: 108746, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34450185

RESUMEN

PURPOSE: To develop a model of focal injury by blue light-emitting diode (LED)-induced phototoxicity (LIP) in pigmented mouse retinas and to study the effects on cone, Iba-1+ cells and retinal pigment epithelium (RPE) cell populations after administration of basic fibroblast growth factor (bFGF) and minocycline, alone or combined. METHODS: In anesthetized dark-adapted adult female pigmented C57BL/6 mice, left pupils were dilated and the eye exposed to LIP (500 lux, 45 s). The retina was monitored longitudinally in vivo with SD-OCT for 7 days (d). Ex vivo, the effects of LIP and its protection with bFGF (0.5 µg) administered alone or combined with minocycline (45 mg/kg) were studied in immunolabeled arrestin-cone outer segments (a+OS) and quantified within a predetermined fixed-size circular area (PCA) centered on the lesion in flattened retinas at 1, 3, 5 or 7d. Moreover, Iba-1+ cells and RPE cell morphology were analysed with Iba-1 and ZO-1 antibodies, respectively. RESULTS: LIP caused a focal lesion within the superior-temporal retina with retinal thinning, particularly the outer retinal layers (116.5 ± 2.9 µm to 36.8 ± 6.3 µm at 7d), and with progressive diminution of a+OS within the PCA reaching minimum values at 7d (6218 ± 342 to 3966 ± 311). Administration of bFGF alone (4519 ± 320) or in combination with minocycline (4882 ± 446) had a significant effect on a+OS survival at 7d and Iba-1+ cell activation was attenuated in the groups treated with minocycline. In parallel, the RPE cell integrity was progressively altered after LIP and administration of neuroprotective components had no restorative effect at 7d. CONCLUSIONS: LIP resulted in progressive outer retinal damage affecting the OS cone population and RPE. Administration of bFGF increased a+OS survival but did not prevent RPE deterioration.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/uso terapéutico , Luz/efectos adversos , Traumatismos Experimentales por Radiación/etiología , Células Fotorreceptoras Retinianas Conos/efectos de la radiación , Degeneración Retiniana/etiología , Animales , Arrestinas/metabolismo , Proteínas de Unión al Calcio/metabolismo , Modelos Animales de Enfermedad , Quimioterapia Combinada , Femenino , Inyecciones Intravítreas , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Microscopía Fluorescente , Minociclina/uso terapéutico , Traumatismos Experimentales por Radiación/diagnóstico por imagen , Traumatismos Experimentales por Radiación/prevención & control , Degeneración Retiniana/diagnóstico por imagen , Degeneración Retiniana/prevención & control , Epitelio Pigmentado de la Retina/metabolismo , Tomografía de Coherencia Óptica
20.
Int J Mol Sci ; 22(13)2021 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-34209135

RESUMEN

Radiation-induced damage to normal lung parenchyma remains a dose-limiting factor in thorax-associated radiotherapy (RT). Severe early and late complications with lungs can increase the risk of morbidity in cancer patients after RT. Herein, senescence of lung epithelial cells following RT-induced cellular stress, or more precisely the respective altered secretory profile, the senescence-associated secretory phenotype (SASP), was suggested as a central process for the initiation and progression of pneumonitis and pulmonary fibrosis. We previously reported that abrogation of certain aspects of the secretome of senescent lung cells, in particular, signaling inhibition of the SASP-factor Ccl2/Mcp1 mediated radioprotection especially by limiting endothelial dysfunction. Here, we investigated the therapeutic potential of a combined metformin treatment to protect normal lung tissue from RT-induced senescence and associated lung injury using a preclinical mouse model of radiation-induced pneumopathy. Metformin treatment efficiently limited RT-induced senescence and SASP expression levels, thereby limiting vascular dysfunctions, namely increased vascular permeability associated with increased extravasation of circulating immune and tumor cells early after irradiation (acute effects). Complementary in vitro studies using normal lung epithelial cell lines confirmed the senescence-limiting effect of metformin following RT finally resulting in radioprotection, while fostering RT-induced cellular stress of cultured malignant epithelial cells accounting for radiosensitization. The radioprotective action of metformin for normal lung tissue without simultaneous protection or preferable radiosensitization of tumor tissue might increase tumor control probabilities and survival because higher radiation doses could be used.


Asunto(s)
Bronquios , Células Epiteliales , Metformina/farmacología , Traumatismos Experimentales por Radiación , Protectores contra Radiación/farmacología , Animales , Bronquios/metabolismo , Bronquios/patología , Senescencia Celular/efectos de los fármacos , Senescencia Celular/efectos de la radiación , Células Epiteliales/metabolismo , Células Epiteliales/patología , Ratones , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/patología , Traumatismos Experimentales por Radiación/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA