Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 757
Filtrar
1.
Nat Commun ; 15(1): 3018, 2024 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-38589357

RESUMO

Ionizing radiation induces cell death in the gastrointestinal (GI) epithelium by activating p53. However, p53 also prevents animal lethality caused by radiation-induced acute GI syndrome. Through single-cell RNA-sequencing of the irradiated mouse small intestine, we find that p53 target genes are specifically enriched in regenerating epithelial cells that undergo fetal-like reversion, including revival stem cells (revSCs) that promote animal survival after severe damage of the GI tract. Accordingly, in mice with p53 deleted specifically in the GI epithelium, ionizing radiation fails to induce fetal-like revSCs. Using intestinal organoids, we show that transient p53 expression is required for the induction of revival stem cells and is controlled by an Mdm2-mediated negative feedback loop. Together, our findings reveal that p53 suppresses severe radiation-induced GI injury by promoting fetal-like reprogramming of irradiated intestinal epithelial cells.


Assuntos
Lesões por Radiação , Proteína Supressora de Tumor p53 , Camundongos , Animais , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Intestinos , Trato Gastrointestinal/metabolismo , Lesões por Radiação/genética , Lesões por Radiação/metabolismo , Células-Tronco/metabolismo , Apoptose/genética
2.
Int J Mol Sci ; 25(5)2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38474123

RESUMO

Radiotherapy-induced cardiac toxicity and consequent diseases still represent potential severe late complications for many cancer survivors who undergo therapeutic thoracic irradiation. We aimed to assess the phenotypic and paracrine features of resident cardiac mesenchymal stromal cells (CMSCs) at early follow-up after the end of thoracic irradiation of the heart as an early sign and/or mechanism of cardiac toxicity anticipating late organ dysfunction. Resident CMSCs were isolated from a rat model of fractionated thoracic irradiation with accurate and clinically relevant heart dosimetry that developed delayed dose-dependent cardiac dysfunction after 1 year. Cells were isolated 6 and 12 weeks after the end of radiotherapy and fully characterized at the transcriptional, paracrine, and functional levels. CMSCs displayed several altered features in a dose- and time-dependent trend, with the most impaired characteristics observed in those exposed in situ to the highest radiation dose with time. In particular, altered features included impaired cell migration and 3D growth and a and significant association of transcriptomic data with GO terms related to altered cytokine and growth factor signaling. Indeed, the altered paracrine profile of CMSCs derived from the group at the highest dose at the 12-week follow-up gave significantly reduced angiogenic support to endothelial cells and polarized macrophages toward a pro-inflammatory profile. Data collected in a clinically relevant rat model of heart irradiation simulating thoracic radiotherapy suggest that early paracrine and transcriptional alterations of the cardiac stroma may represent a dose- and time-dependent biological substrate for the delayed cardiac dysfunction phenotype observed in vivo.


Assuntos
Cardiopatias , Células-Tronco Mesenquimais , Lesões por Radiação , Ratos , Humanos , Animais , Cardiotoxicidade/metabolismo , Células Endoteliais/metabolismo , Células-Tronco Mesenquimais/metabolismo , Fenótipo , Cardiopatias/metabolismo , Lesões por Radiação/metabolismo
3.
Int J Mol Sci ; 25(5)2024 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-38473879

RESUMO

Pelvic cancer survivors who were treated with radiation therapy are at risk for developing (hemorrhagic) radiation cystitis (RC) many years after completion of radiation therapy. Patients with RC suffer from lower urinary tract symptoms, including frequency, nocturia, pelvic pain, and incontinence. In advanced stages, hematuria can occur, potentially escalating to life-threatening levels. Current therapeutic options for RC are limited, partly due to ethical concerns regarding bladder biopsy in patients with fragile bladder tissue. This study aimed to leverage our established preclinical model to elucidate the molecular pathways implicated in radiation-induced tissue changes in the bladder. Female C57Bl/6 mice received a single dose of 40 Gy using CT-guided imaging and a two-beam irradiation approach using the SARRP irradiator. Bladders from irradiated and age-matched littermate controls were harvested at 1 week [n = 5/group] or 6 months [n = 5/group] after irradiation, RNA was harvested, and mRNA sequencing was performed at paired-end 150bp on the Illumina NovaSeq6000 with a target of 30 million reads per sample. Following RNA sequencing, thorough bioinformatics analysis was performed using iPathwayGuide v2012 (ADVAITA Bioinformatics). Findings of the RNA sequencing were validated using qPCR analysis. At 1 week post-irradiation, altered gene expression was detected in genes involved in DNA damage response, apoptosis, and transcriptional regulation. By 6 months post-irradiation, significant changes in gene expression were observed in inflammation, collagen catabolism, and vascular health. Affected pathways included the p53, JAK-STAT, and PI3K-Akt pathways. These findings were validated in vivo in bladder tissues from our preclinical model. This is the first study to determine the molecular changes in the bladder in response to radiation treatment. We have successfully pinpointed several pathways and specific genes that undergo modification, thereby contributing to the progression of radiation cystitis. These insights enhance our understanding of the pathophysiology of radiation cystitis and may ultimately pave the way to the identification of potential new therapeutic targets.


Assuntos
Cistite , Lesões por Radiação , Camundongos , Animais , Humanos , Feminino , Recém-Nascido , Fosfatidilinositol 3-Quinases/metabolismo , Cistite/patologia , Bexiga Urinária/patologia , Lesões por Radiação/metabolismo , Análise de Sequência de RNA
4.
Life Sci ; 341: 122486, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38331314

RESUMO

PURPOSE: Damage to the hematopoietic system and functional inhibition are severe consequences of radiation exposure. In this study, we have investigated the effect of empagliflozin on radiation-induced hematopoietic damage, with the aim of providing new preventive approach to such injuries. METHODS AND MATERIALS: Mice were given 4 Gy total body irradiation (TBI) 1 h after the oral administration of empagliflozin, followed by the continuous administration of the same dose of empagliflozin for 6d, and then sacrificed on the 10th day after irradiation. The reactive oxygen species (ROS) levels in hematopoietic cells and their regulatory mechanisms were also been investigated. Colony forming unit granulocyte macrophage assay and bone marrow transplantation assays were performed to detect the function of the bone marrow cells. KEY FINDINGS: Empagliflozin increased the cell viability, reduced ROS levels, and attenuated apoptosis in vitro after the bone marrow cells were exposed to 1 Gy radiation. Empagliflozin significantly attenuated ionizing radiation injuries to the hematopoietic system, increased the peripheral blood cell count, and enhanced the proportion and function of hematopoietic stem cells in mice exposed to 4 Gy TBI. These effects may be related to the NOX-4/ROS/p38 pathway-mediated suppression of MAPK in hematopoietic stem cells. Empagliflozin also influenced the expression of Nrf-2 and increased glutathione peroxidase activity, thereby promoting the clearance of reactive oxygen species. Furthermore, empagliflozin mitigated metabolic abnormalities by inhibiting the mammalian target of rapamycin. SIGNIFICANCE: Our study has demonstrated that empagliflozin can reduce radiation-induced injury in hematopoietic stem cells. This finding suggests that empagliflozin is a promising novel agent for preventing radiation-induced damage to the hematopoietic system.


Assuntos
Glucosídeos , Células-Tronco Hematopoéticas , Lesões por Radiação , Animais , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Ensaio de Unidades Formadoras de Colônias , Compostos Benzidrílicos/farmacologia , Compostos Benzidrílicos/metabolismo , Lesões por Radiação/metabolismo , Irradiação Corporal Total , Camundongos Endogâmicos C57BL , Mamíferos/metabolismo
5.
Nat Commun ; 15(1): 137, 2024 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-38167344

RESUMO

Radiation-induced gastrointestinal syndrome is a major complication and limiting factor for radiotherapy. Tumor suppressor p53 has a protective role in radiation-induced gastrointestinal toxicity. However, its underlying mechanism remains unclear. Here we report that regulating the IL12-p40/MHC class II signaling pathway is a critical mechanism by which p53 protects against radiation-induced gastrointestinal syndrome. p53 inhibits the expression of inflammatory cytokine IL12-p40, which in turn suppresses the expression of MHC class II on intestinal epithelial cells to suppress T cell activation and inflammation post-irradiation that causes intestinal stem cell damage. Anti-IL12-p40 neutralizing antibody inhibits inflammation and rescues the defects in intestinal epithelial regeneration post-irradiation in p53-deficient mice and prolongs mouse survival. These results uncover that the IL12-p40/MHC class II signaling mediates the essential role of p53 in ensuring intestinal stem cell function and proper immune reaction in response to radiation to protect mucosal epithelium, and suggest a potential therapeutic strategy to protect against radiation-induced gastrointestinal syndrome.


Assuntos
Lesões por Radiação , Proteína Supressora de Tumor p53 , Animais , Camundongos , Proteína Supressora de Tumor p53/metabolismo , Apoptose/efeitos da radiação , Mucosa Intestinal/metabolismo , Lesões por Radiação/metabolismo , Inflamação/metabolismo , Interleucina-12/metabolismo
6.
Radiat Res ; 201(2): 160-173, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38124379

RESUMO

The effect of ionizing radiation on the gastrointestinal tract is a common complication of abdominal and pelvic radiotherapy. However, the pathological features of radiation enteropathy and its effective medical intervention regimen is still a global challenge. Here, we explored the role and mechanism of enteric alpha-defensins (EαDs) in protecting against radiation enteropathy. To address this, we utilized EαDs-deficiency mice, in which the matrix metallopeptidase 7 to activate Paneth cell α-defensins was knockout (KO) mice, and the complementary wild-type (WT) control mice for this study. Remarkably, the KO mice were more susceptible to 5.0 Gy total-body irradiation, resulting in worse clinic scores and lower survival rate, compared with the wild-type mice. Histological examination indicated that the KO mice were subjected to slow recovery of intestinal villus and mucosa function, characterized by the reduced expression of TFF3, Glut1 and Muc2. In addition, compared with the wild-type controls, the KO mice experienced serious inflammation response in intestinal tissue, indicated by the remarkably increased expression level of IL-1ß, IL-6 and IL-12. Using high-throughput sequencing analysis, we found that the intestinal bacterial community of the KO mice was more prone to dysbiosis than that of the WT mice, with significantly increased abundance of opportunistic pathogenic bacteria, such as Streptococcus sp. and Escherichia-Shigella sp., whereas remarkably decreased probiotics harboring Lactobacillus sp., Desulfovibrio sp. etc. Fecal metabolomics analysis indicated that the relative abundance of 31 metabolites arose significantly different between WT and KO mice on day 10 after radiation exposure. A subset of differential metabolites to regulate host metabolism and immunity, such as acetic acid, acetate, butanoic acid, was negatively correlated with the alteration of gut microbiota in the irradiated KO mice. This study provides new insight into EαDs contribution to the recovery of radiation-induced intestinal damage, and suggests a potential novel target to prevent the adverse effects of radiotherapy.


Assuntos
Microbioma Gastrointestinal , Lesões por Radiação , alfa-Defensinas , Camundongos , Animais , alfa-Defensinas/genética , alfa-Defensinas/metabolismo , Microbioma Gastrointestinal/efeitos da radiação , Intestinos , Mucosa Intestinal/metabolismo , Fezes/microbiologia , Lesões por Radiação/metabolismo , Camundongos Knockout , Camundongos Endogâmicos C57BL
7.
Commun Biol ; 6(1): 1248, 2023 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-38071238

RESUMO

Intestine is a highly radiation-sensitive organ that could be injured during the radiotherapy for pelvic, abdominal, and retroperitoneal tumors. However, the dynamic change of the intestinal microenvironment related to radiation-induced intestine injury (RIII) is still unclear. Using single-cell RNA sequencing, we pictured a dynamic landscape of the intestinal microenvironment during RIII and regeneration. We showed that the various cell types of intestine exhibited heterogeneous radiosensitivities. We revealed the distinct dynamic patterns of three subtypes of intestinal stem cells (ISCs), and the cellular trajectory analysis suggested a complex interconversion pattern among them. For the immune cells, we found that Ly6c+ monocytes can give rise to both pro-inflammatory macrophages and resident macrophages after RIII. Through cellular communication analysis, we identified a positive feedback loop between the macrophages and endothelial cells, which could amplify the inflammatory response induced by radiation. Besides, we identified different T cell subtypes and revealed their role in immunomodulation during the early stage of RIII through inflammation and defense response relevant signaling pathways. Overall, our study provides a valuable single-cell map of the multicellular dynamics during RIII and regeneration, which may facilitate the understanding of the mechanism of RIII.


Assuntos
Enteropatias , Lesões por Radiação , Humanos , Células Endoteliais/patologia , Intestinos/patologia , Lesões por Radiação/metabolismo , Células-Tronco/metabolismo , Microambiente Celular
8.
Int J Mol Sci ; 24(24)2023 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-38139042

RESUMO

Radiotherapy (RT) is one of three major treatments for malignant tumors, and one of its most common side effects is skin and soft tissue injury. However, the treatment of these remains challenging. Several studies have shown that mesenchymal stem cell (MSC) treatment enhances skin wound healing. In this study, we extracted human dermal fibroblasts (HDFs) and adipose-derived stem cells (ADSCs) from patients and generated an in vitro radiation-induced skin injury model with HDFs to verify the effect of conditioned medium derived from adipose-derived stem cells (ADSC-CM) and extracellular vesicles derived from adipose-derived stem cells (ADSC-EVs) on the healing of radiation-induced skin injury. The results showed that collagen synthesis was significantly increased in wounds treated with ADSC-CM or ADSC-EVs compared with the control group, which promoted the expression of collagen-related genes and suppressed the expression of inflammation-related genes. These findings indicated that treatment with ADSC-CM or ADSC-EVs suppressed inflammation and promoted extracellular matrix deposition; treatment with ADSC-EVs also promoted fibroblast proliferation. In conclusion, these results demonstrate the effectiveness of ADSC-CM and ADSC-EVs in the healing of radiation-induced skin injury.


Assuntos
Vesículas Extracelulares , Lesões por Radiação , Humanos , Meios de Cultivo Condicionados/farmacologia , Meios de Cultivo Condicionados/metabolismo , Tecido Adiposo/metabolismo , Células-Tronco/metabolismo , Lesões por Radiação/metabolismo , Inflamação/metabolismo , Colágeno/metabolismo
9.
Int J Mol Sci ; 24(22)2023 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-38003456

RESUMO

Radiation-induced lung fibrosis (RILF) is a common complication of radiotherapy in lung cancer. However, to date no effective treatment has been developed for this condition. NXC736 is a novel small-molecule compound that inhibits NLRP3, but its effect on RILF is unknown. NLRP3 activation is an important trigger for the development of RILF. Thus, we aimed to evaluate the therapeutic effect of NXC736 on lung fibrosis inhibition using a RILF animal model and to elucidate its molecular signaling pathway. The left lungs of mice were irradiated with a single dose of 75 Gy. We observed that NXC736 treatment inhibited collagen deposition and inflammatory cell infiltration in irradiated mouse lung tissues. The damaged lung volume, evaluated by magnetic resonance imaging, was lower in NXC736-treated mice than in irradiated mice. NXC736-treated mice exhibited significant changes in lung function parameters. NXC736 inhibited inflammasome activation by interfering with the NLRP3-ASC-cleaved caspase-1 interaction, thereby reducing the expression of IL-1ß and blocking the fibrotic pathway. In addition, NXC736 treatment reduced the expression of epithelial-mesenchymal transition markers such as α-SMA, vimentin, and twist by blocking the Smad 2,3,4 signaling pathway. These data suggested that NXC736 is a potent therapeutic agent against RILF.


Assuntos
Fibrose Pulmonar , Lesões por Radiação , Camundongos , Animais , Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/etiologia , Fibrose Pulmonar/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Pulmão/patologia , Fibrose , Inflamassomos/metabolismo , Lesões por Radiação/metabolismo , Transdução de Sinais , Síndrome da Fibrose por Radiação
10.
Food Res Int ; 174(Pt 1): 113582, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37986451

RESUMO

Radiation esophagitis (RE) is an inimical event that requires proper management while carrying out radiotherapy for thoracic cancers. The present study investigates the protective effect of dry fruits of the culinary and folkloric spice Amomum subulatum against experimental thoracic radiation-induced esophagitis. C57BL/6 mice were subjected to 25 Gy whole thorax irradiation and administered with 250 mg/kg body weight of methanolic extract of A. subulatum dry fruits (MEAS) orally for four consecutive weeks. Changes in tissue antioxidant activities, oxidative stress parameters, expression of antioxidant, inflammation, and fibrosis-related genes were observed. Administration of MEAS boosted antioxidant status, thereby reducing radiation-induced oxidative stress in the esophagus. PCR (polymerase chain reaction) results showed decreased expression of apoptosis, inflammation, and fibrosis-associated genes as well as increased expression of vital cytoprotective and antioxidant genes in MEAS-treated mice, manifesting its protective effect against radiation-induced oxidative stress, inflammatory responses, and fibrosis in the esophagus. Further, histopathology, immunohistochemistry (Cyclooxygenase-2), and Masson's Trichrome staining ascertained the protective effect of MEAS in alleviating radiation-induced esophageal injury. The synergistic effect of bioactive phytochemicals in MEAS with potent antioxidant and anti-inflammatory efficacies might have contributed to its mitigating effect against RE. Taken together, our results ascertained the radioprotective potential of MEAS, suggesting its possible nutraceutical application as a radiation countermeasure.


Assuntos
Amomum , Esofagite , Lesões por Radiação , Camundongos , Animais , Antioxidantes/farmacologia , Frutas/metabolismo , Camundongos Endogâmicos C57BL , Lesões por Radiação/prevenção & controle , Lesões por Radiação/metabolismo , Esofagite/prevenção & controle , Esofagite/metabolismo , Inflamação/prevenção & controle , Fibrose
11.
Radiat Res ; 200(5): 462-473, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37796808

RESUMO

The effect of autophagy on the radiation-induced bystander effect (RIBE) in vivo is unclear. Here, the whole brains of microtubule-associated protein 1A/1B-light chain 3 (LC3) and C57BL/6 (B6) mice were irradiated once (10 Gy)(IR1), given 3 fractions in three weeks (IR3), or 6 fractions in six weeks (IR6). The median survival of LC3 mice was 56.5 days, and that of B6 mice was 65 days after IR6. LC3 mice showed more congestion and fibrosis in the lung after the IR3 and IR6 irradiation protocols than B6 mice. Quantitative proteomics of serum samples and lung RNA sequencing of the LC3 group showed that the common most clustered pathway of the IR3 group was the elastic fiber formation pathway, which contained Periostin (POSTN). POSTN in the motoneurons increased with increasing number of radiation fractions in LC3 mice. A 1 µg/g POSTN neutralizing antibody reduced the lung fibrosis of LC3 mice exposed to IR3 by one-third, and significantly prolonged the survival time of LC3 mice exposed to IR6. LDN-214117 and LRRK2-in-1 were the best two of sixteen transforming growth factor-beta1 (TGF-ß) receptor and autophagy mediators to decrease Postn mRNA. These data led us to conclude that LC3 accelerated motoneuron secretion of POSTN and aggravated the RIBE in the lung after brain irradiation.


Assuntos
Fibrose Pulmonar , Lesões por Radiação , Camundongos , Animais , Camundongos Endogâmicos C57BL , Pulmão/efeitos da radiação , Fibrose Pulmonar/metabolismo , Lesões por Radiação/metabolismo , Encéfalo , Neurônios Motores
12.
Cells ; 12(19)2023 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-37830619

RESUMO

GADD45a is a gene we previously reported as a mediator of responses to acute lung injury. GADD45a-/- mice express decreased Akt and increased Akt ubiquitination due to the reduced expression of UCHL1 (ubiquitin c-terminal hydrolase L1), a deubiquitinating enzyme, while GADD45a-/- mice have increased their susceptibility to radiation-induced lung injury (RILI). Separately, we have reported a role for sphingolipids in RILI, evidenced by the increased RILI susceptibility of SphK1-/- (sphingosine kinase 1) mice. A mechanistic link between UCHL1 and sphingolipid signaling in RILI is suggested by the known polyubiquitination of SphK1. Thus, we hypothesized that the regulation of SphK1 ubiquitination by UCHL1 mediates RILI. Initially, human lung endothelial cells (EC) subjected to radiation demonstrated a significant upregulation of UCHL1 and SphK1. The ubiquitination of EC SphK1 after radiation was confirmed via the immunoprecipitation of SphK1 and Western blotting for ubiquitin. Further, EC transfected with siRNA specifically for UCHL1 or pretreated with LDN-5744, as a UCHL1 inhibitor, prior to radiation were noted to have decreased ubiquitinated SphK1 in both conditions. Further, the inhibition of UCHL1 attenuated sphingolipid-mediated EC barrier enhancement was measured by transendothelial electrical resistance. Finally, LDN pretreatment significantly augmented murine RILI severity. Our data support the fact that the regulation of SphK1 expression after radiation is mediated by UCHL1. The modulation of UCHL1 affecting sphingolipid signaling may represent a novel RILI therapeutic strategy.


Assuntos
Lesão Pulmonar , Lesões por Radiação , Camundongos , Humanos , Animais , Lesão Pulmonar/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células Endoteliais/metabolismo , Ubiquitina Tiolesterase/genética , Ubiquitina Tiolesterase/metabolismo , Pulmão/metabolismo , Ubiquitina/metabolismo , Esfingolipídeos/metabolismo , Lesões por Radiação/metabolismo
13.
J Cell Mol Med ; 27(23): 3839-3850, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37723905

RESUMO

Radiation-induced lung injury (RILI), divided into early radiation pneumonia (RP) and late radiation-induced pulmonary fibrosis (RIPF), is a common serious disease after clinical chest radiotherapy or nuclear accident, which seriously threatens the life safety of patients. There has been no effective prevention or treatment strategy till now. Epithelial-mesenchymal transition (EMT) is a key step in the occurrence and development of RILI. In this study, we demonstrated that emetine dihydrochloride (EDD) alleviated RILI through inhibiting EMT. We found that EDD significantly attenuated EMT-related markers, reduced Smad3 phosphorylation expression after radiation. Then, for the first time, we observed EDD alleviated lung hyperaemia and reduced collagen deposit induced by irradiation, providing protection against RILI. Finally, it was found that EDD inhibited radiation-induced EMT in lung tissues. Our study suggested that EDD alleviated RILI through inhibiting EMT by blocking Smad3 signalling pathways. In summary, our results indicated that EDD is a novel potential radioprotector for RILI.


Assuntos
Lesão Pulmonar , Fibrose Pulmonar , Lesões por Radiação , Humanos , Lesão Pulmonar/tratamento farmacológico , Lesão Pulmonar/etiologia , Lesão Pulmonar/metabolismo , Emetina/farmacologia , Pulmão/patologia , Lesões por Radiação/metabolismo , Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/etiologia , Fibrose Pulmonar/metabolismo , Transição Epitelial-Mesenquimal
14.
Cell Biochem Funct ; 41(8): 1115-1132, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37653677

RESUMO

The biogenic synthesis of nanoparticles has drawn significant attention. The spleen is the largest lymphatic organ that is adversely impacted during irradiation. The current study was designated to evaluate the possible anti-inflammatory effect of matcha-silver nanoparticles (M-AgNPs) to reduce inflammation associated with γ-radiation induced-oxidative stress and inflammation in rats' spleen. Silver nanoparticles (AgNPs) were synthesized by biogenic synthesis using a green sonochemical method from matcha (M) green tea. The obtained M-AgNPs were extensively characterized by dynamic light scattering, transmission electron microscopy, thermogravimetric analysis, and Fourier-transform infrared spectroscopy. Using zetasizer analysis, the surface charge, particle size, and radical scavenging DPPH assay of M-AgNPs were also examined. Biocompatibility and cytotoxicity were analyzed by MTT assay, and the IC50 was calculated. Four groups of 24 Wistar rats each had an equal number of animals. The next step involved measuring the levels of oxidative stress markers in the rat splenic tissue. Additionally, the amounts of inflammatory protein expression were evaluated using the ELISA analysis. The results indicated the formation of spherical nanoparticles of pure Ag° coated with matcha polyphenols at the nanoscale, as well as uniform monodisperse particles suited for cellular absorption. Results revealed that M-AgNPs improved all biochemical parameters. Furthermore, M-AgNPs relieve inflammation by reducing the expression of NOD-like receptor family pyrin domain-containing 3 (NLRP3), interleukin-1ß (IL-1ß), and enhancing the levels of ileSnt information regulator 1 (SIRT1). Histopathological examinations demonstrated the ability of M-AgNPs to overcome the damage consequent to irradiation and recover the spleen's cellular structure. These results confirmed that matcha is a potential biomaterial for synthesizing AgNPs, which can be exploited for their anti-inflammatory activity.


Assuntos
Nanopartículas Metálicas , Prata , Animais , Ratos , Anti-Inflamatórios , Raios gama , Inflamação/tratamento farmacológico , Inflamação/patologia , Nanopartículas Metálicas/química , Estresse Oxidativo , Ratos Wistar , Transdução de Sinais , Prata/farmacologia , Prata/química , Prata/uso terapêutico , Sirtuína 1 , Baço , Lesões por Radiação/genética , Lesões por Radiação/metabolismo , Lesões por Radiação/prevenção & controle , Chá
15.
Int J Radiat Biol ; 99(12): 1865-1878, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37531370

RESUMO

PURPOSE: Radiation-induced gastrointestinal injury (RIGI) is a serious side effect of abdominal and pelvic radiotherapy, which often limits the treatment of gastrointestinal and gynaecological cancers. RIGI is also observed during accidental radiological or nuclear scenarios with no approved agents available till date to prevent or mitigate RIGI in humans. Trichostatin A (TSA), an epigenetic modulator, has been currently in clinical trials for cancer treatment and is also well known for its antibiotic and antifungal properties. METHODS: In this study, partial body (abdominal) irradiation mice model was used to investigate the mitigative effect of TSA against gastrointestinal toxicity caused by gamma radiation. Mice were checked for alterations in mean body weight, diarrheal incidence, disease activity index and survival against 15 Gy radiation. Structural abnormalities in intestine and changes in microbiota composition were studied by histopathology and 16S rRNA sequencing of fecal samples respectively. Immunoblotting and biochemical assays were performed to check protein nitrosylation, expression of inflammatory mediators, infiltration of inflammatory cells and changes in pro-inflammatory cytokine. RESULTS: TSA administration to C57Bl/6 mice improved radiation induced mean body weight loss, maintained better health score, reduced disease activity index and promoted survival. The 16S rRNA sequencing of fecal DNA demonstrated that TSA influenced the fecal microbiota dynamics with significant alterations in the Firmicutes/Bacteriodetes ratio. TSA effectively mitigated intestinal injury, down-regulated NF-κB, Cox-2, iNOS expression, inhibited PGE2 and protein nitrosylation levels in irradiated intestine. The upregulation of NLRP3-inflammasome complex and infiltrations of inflammatory cells in the inflamed intestine were also prevented by TSA. Subsequently, the myeloperoxidase activity in intestine alongwith serum IL-18 levels was found reduced. CONCLUSION: These findings provide evidence that TSA inhibits inflammatory mediators, alleviates gut dysbiosis, and promotes structural restoration of the irradiated intestine. TSA, therefore, can be considered as a potential agent for mitigation of RIGI in humans.


Assuntos
Microbioma Gastrointestinal , Lesões por Radiação , Humanos , Animais , Camundongos , Microbioma Gastrointestinal/efeitos da radiação , RNA Ribossômico 16S/genética , Lesões por Radiação/tratamento farmacológico , Lesões por Radiação/metabolismo , Anti-Inflamatórios , Mediadores da Inflamação , Camundongos Endogâmicos C57BL
16.
Zhongguo Fei Ai Za Zhi ; 26(6): 467-472, 2023 Jun 20.
Artigo em Chinês | MEDLINE | ID: mdl-37488084

RESUMO

Radiation therapy is one of the main treatment methods for patients with thoracic malignant tumors, which can effectively improve the survival rate of the patients. However, radiation therapy can also cause damage to normal tissues while treating tumors, leading to radiation-induced lung injury such as radiation pneumonia and pulmonary fibrosis. Radiation-induced lung injury is a complex pathophysiological process involving many factors, and its prevention and treatment is one of the difficult problems in the field of radiation medicine. Therefore, the search for sensitive predictors of radiation-induced lung injury can guide clinical radiotherapy and reduce the incidence of radiation-induced lung injury. With the in-depth study of intestinal flora, it can drive immune cells or metabolites to reach lung tissue through the circulatory system to play a role, and participate in the occurrence, development and treatment of lung diseases. At present, there are few studies on intestinal flora and radiation-induced lung injury. Therefore, this paper will comprehensively elaborate the interaction between intestinal flora and radiation-induced lung injury, so as to provide a new direction and strategy for studying the protective effect of intestinal flora on radiation-induced lung injury.
.


Assuntos
Microbioma Gastrointestinal , Lesão Pulmonar , Neoplasias Pulmonares , Lesões por Radiação , Neoplasias Torácicas , Humanos , Lesão Pulmonar/etiologia , Lesão Pulmonar/prevenção & controle , Neoplasias Pulmonares/radioterapia , Pulmão/patologia , Lesões por Radiação/complicações , Lesões por Radiação/metabolismo
17.
Radiat Res ; 200(2): 151-161, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37327123

RESUMO

Radiation-induced esophageal injury (RIEI) is a major dose-limiting complication of radiotherapy, mainly acute esophagitis. However, understanding of radiation injury and repair mechanisms in esophageal epithelial cells remains limited. MiR-132-3p and its uridylated isoform (miR-132-3p-UUU) are upregulated in radiation esophageal injury, yet their role in radiation-induced esophageal injury progression remains unexplored. We expressed miR-132-3p and its uridine form in irradiated human esophageal epithelial cells (HEEC) and secreted exosomes was examined by real-time polymerase chain reaction (RT-PCR). Cell proliferation, migration, apoptosis and colony formation were used to determine biological effects. Cell cycle assays and dual luciferase reporter assays were used to assess the relationship between miR-132-3p and its uridylated isoforms and MEF2A. The addition of miR-132-3p mimics or overexpression of miR-132-3p significantly inhibited the proliferation and migration of esophageal epithelial cells (HEEC cells as well as primary cells) and increased radiation damage. This was reversed by its uridylated isoform by reducing binding to MEF2A and regulating the cell cycle. Furthermore, miR-132-3p and its triuridylated isomer also regulate apoptosis after irradiation through pathways other than reactive oxygen species (ROS). In conclusion, our data reveal that radiation-induced miR-132-3p uridylation and exosome-mediated intercellular communication and tri-uridylated isoforms are protective against radiation-induced esophageal injury. Furthermore, miR-132-3p offers new opportunities as a promising biomarker widely present in human body fluids for the prediction of radiation esophagitis as a biomarker.


Assuntos
Esofagite , Exossomos , MicroRNAs , Lesões por Radiação , Humanos , Apoptose , Biomarcadores/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Esofagite/metabolismo , Esofagite/patologia , Exossomos/metabolismo , MicroRNAs/genética , Lesões por Radiação/metabolismo
18.
Front Immunol ; 14: 1151250, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37168858

RESUMO

Introduction: Exposure to high-dose ionizing radiation causes tissue injury, infections and even death due to immune dysfunction. The triggering receptor expressed on myeloid cells-1 (TREM-1) has been demonstrated to critically amplify and dysregulate immune responses. However, the role of TREM-1 in radiation injury remains unknown. Extracellular cold-inducible RNA-binding protein (eCIRP), a new damage-associated molecular pattern, is released from activated or stressed cells during inflammation. We hypothesized that ionizing radiation upregulates TREM-1 expression via eCIRP release to worsen survival. Methods: RAW264.7 cells and peritoneal macrophages collected from C57BL/6 wild-type (WT) mice were exposed to 5- and 10-Gray (Gy) radiation. C57BL/6 WT and CIRP-/- mice underwent 10-Gy total body irradiation (TBI). TREM-1 expression on RAW264.7 cells and peritoneal macrophages in vitro and in vivo were evaluated by flow cytometry. eCIRP levels in cell culture supernatants and in peritoneal lavage isolated from irradiated mice were evaluated by Western blotting. We also evaluated 30-day survival in C57BL/6 WT, CIRP-/- and TREM-1-/- mice after 6.5-Gy TBI. Results: The surface protein and mRNA levels of TREM-1 in RAW264.7 cells were significantly increased at 24 h after 5- and 10-Gy radiation exposure. TREM-1 expression on peritoneal macrophages was significantly increased after radiation exposure in vitro and in vivo. eCIRP levels were significantly increased after radiation exposure in cell culture supernatants of peritoneal macrophages in vitro and in peritoneal lavage in vivo. Moreover, CIRP-/- mice exhibited increased survival after 6.5-Gy TBI compared to WT mice. Interestingly, TREM-1 expression on peritoneal macrophages in CIRP-/- mice was significantly decreased compared to that in WT mice at 24 h after 10-Gy TBI. Furthermore, 30-day survival in TREM-1-/- mice was significantly increased to 64% compared to 20% in WT mice after 6.5-Gy TBI. Conclusion: Our data indicate that ionizing radiation increases TREM-1 expression in macrophages via the release of eCIRP, and TREM-1 contributes to worse survival after total body irradiation. Thus, targeting TREM-1 could have the potential to be developed as a novel medical countermeasure for radiation injury.


Assuntos
Macrófagos , Lesões por Radiação , Animais , Camundongos , Inflamação/metabolismo , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Lesões por Radiação/genética , Lesões por Radiação/metabolismo , Receptor Gatilho 1 Expresso em Células Mieloides/genética , Receptor Gatilho 1 Expresso em Células Mieloides/metabolismo
19.
Int J Mol Sci ; 24(2)2023 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-36675266

RESUMO

The damaging effect of ionizing radiation (IR) exposure results in the disturbance of the gut natural barrier, followed by the development of severe gastrointestinal injury. However, the dose and application segment are known to determine the effects of IR. In this study, we demonstrated the dose- and segment-specificity of tight junction (TJ) alteration in IR-induced gastrointestinal injury in rats. Male Wistar rats were subjected to a total-body X-ray irradiation at doses of 2 or 10 Gy. Isolated jejunum and colon segments were tested in an Ussing chamber 72 h after exposure. In the jejunum, 10-Gy IR dramatically altered transepithelial resistance, short-circuit current and permeability for sodium fluorescein. These changes were accompanied by severe disturbance of histological structure and total rearrangement of TJ content (increased content of claudin-1, -2, -3 and -4; multidirectional changes in tricellulin and occludin). In the colon of 10-Gy irradiated rats, lesions of barrier and transport functions were less pronounced, with only claudin-2 and -4 altered among TJ proteins. The 2-Gy IR did not change electrophysiological characteristics or permeability in the colon or jejunum, although slight alterations in jejunum histology were noted, emphasized with claudin-3 increase. Considering that TJ proteins are critical for maintaining epithelial barrier integrity, these findings may have implications for countermeasures in gastrointestinal acute radiation injury.


Assuntos
Lesões por Radiação , Proteínas de Junções Íntimas , Ratos , Masculino , Animais , Proteínas de Junções Íntimas/metabolismo , Mucosa Intestinal/metabolismo , Ratos Wistar , Junções Íntimas/metabolismo , Ocludina/metabolismo , Radiação Ionizante , Lesões por Radiação/metabolismo , Permeabilidade
20.
Rev Environ Health ; 38(2): 295-311, 2023 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-35438851

RESUMO

The gastrointestinal (GI) system has rapidly proliferating and differentiating cells, which make it one of the most radiosensitive organs in the body. Exposure to high dose of ionising radiation (IR) during radiotherapy may generate a variety of reactive oxygen species (ROS) and reactive nitrogen species (RNS) including radicals, cause some side effects such as nausea, vomiting, diarrhoea, pain, ulceration, mal-absorption etc. Irradiation disrupts GI system by damaging proliferating stem cells of the crypts that alters the histology and physiology of intestine. Radiation damage reflects the qualitative and quantitative changes in intestinal epithelial stem cells like enterocytes, enteroendocrine cells, goblet cells and Paneth cells. The damaging effects of radiation to bio-molecules and cellular structures can alter gene signalling cascades and grounds genomic instability, protein modifications, cell senescence and cell death. The signalling pathways of GI tract includes Wnt, BMP, Hedgehog, PTEN/PI3K and Notch plays an important role in self-renewal of intestinal stem cells (ISCs) and maintaining the balance between self-renewal and differentiation of ISCs. Various radiation countermeasures including radioprotectors and mitigators are under development phase globally but still not approved for clinical applications during any radiation emergencies. In view of above, present review highlights cellular and molecular interruptions of GI system due to acute and chronic GI radiation injury, role of radioprotectors in signalling cascade modulations in GI epithelium and involvement of ISC markers in radioprotection.


Assuntos
Lesões por Radiação , Protetores contra Radiação , Humanos , Protetores contra Radiação/farmacologia , Protetores contra Radiação/metabolismo , Intestinos/patologia , Lesões por Radiação/metabolismo , Lesões por Radiação/patologia , Células-Tronco/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...