Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 10.957
Filtrar
1.
Am J Physiol Gastrointest Liver Physiol ; 326(6): G631-G642, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38593468

RESUMEN

Lysophosphatidic acid (LPA) is a bioactive lipid molecule that regulates a wide array of cellular functions, including proliferation, differentiation, and survival, via activation of cognate receptors. The LPA5 receptor is highly expressed in the intestinal epithelium, but its function in restoring intestinal epithelial integrity following injury has not been examined. Here, we use a radiation-induced injury model to study the role of LPA5 in regulating intestinal epithelial regeneration. Control mice (Lpar5f/f) and mice with an inducible, epithelial cell-specific deletion of Lpar5 in the small intestine (Lpar5IECKO) were subjected to 10 Gy total body X-ray irradiation and analyzed during recovery. Repair of the intestinal mucosa was delayed in Lpar5IECKO mice with reduced epithelial proliferation and increased crypt cell apoptosis. These effects were accompanied by reduced numbers of OLFM4+ intestinal stem cells (ISCs). The effects of LPA5 on ISCs were corroborated by studies using organoids derived from Lgr5-lineage tracking reporter mice with deletion of Lpar5 in Lgr5+-stem cells (Lgr5Cont or Lgr5ΔLpar5). Irradiation of organoids resulted in fewer numbers of Lgr5ΔLpar5 organoids retaining Lgr5+-derived progenitor cells compared with Lgr5Cont organoids. Finally, we observed that impaired regeneration in Lpar5IECKO mice was associated with reduced numbers of Paneth cells and decreased expression of Yes-associated protein (YAP), a critical factor for intestinal epithelial repair. Our study highlights a novel role for LPA5 in regeneration of the intestinal epithelium following irradiation and its effect on the maintenance of Paneth cells that support the stem cell niche.NEW & NOTEWORTHY We used mice lacking expression of the lysophosphatidic acid receptor 5 (LPA5) in intestinal epithelial cells and intestinal organoids to show that the LPA5 receptor protects intestinal stem cells and progenitors from radiation-induced injury. We show that LPA5 induces YAP signaling and regulates Paneth cells.


Asunto(s)
Proliferación Celular , Mucosa Intestinal , Receptores del Ácido Lisofosfatídico , Regeneración , Transducción de Señal , Proteínas Señalizadoras YAP , Animales , Receptores del Ácido Lisofosfatídico/metabolismo , Receptores del Ácido Lisofosfatídico/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efectos de la radiación , Ratones , Regeneración/efectos de la radiación , Proteínas Señalizadoras YAP/metabolismo , Proliferación Celular/efectos de la radiación , Células Madre/efectos de la radiación , Células Madre/metabolismo , Organoides/metabolismo , Organoides/efectos de la radiación , Ratones Noqueados , Apoptosis/efectos de la radiación , Lisofosfolípidos/metabolismo , Intestino Delgado/efectos de la radiación , Intestino Delgado/metabolismo , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/patología
2.
Int Immunopharmacol ; 133: 111987, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38652961

RESUMEN

Radiation-induced kidney injury is a common side effect of radiotherapy, as the pelvic region is in close proximity to the kidneys, posing a risk of inducing radiation-induced kidney injury when treating any pelvic malignancies with radiotherapy. This type of injury typically manifests as chronic kidney disease a few months after radiotherapy, with the potential to progress to end-stage renal disease. Radiation-induced damage involves various components of the kidney, including glomeruli, tubules, interstitium, and extracellular matrix. Therefore, investigating its molecular mechanisms is crucial. In this study, we extensively searched literature databases, selecting recent transcriptomic studies related to acute kidney injury (AKI) published in the past decade. We downloaded the raw RNA sequencing datasets GSE30718 and GSE66494 related to AKI from the GEO database and identified that intestinal-type lectin ITLN1 plays a significant role in regulating radiation-induced kidney injury in rats. Differential gene analysis was performed using chip data from the GEO database, and further bioinformatics analysis identified 13 genes that may be involved in regulating kidney injury, with ITLN1 being the most relevant to kidney damage, thus selected as the target gene for this study. Subsequently, a rat model of radiation-induced kidney injury was established for experimental validation, assessing kidney tissue morphology and injury extent through staining observation and immunohistochemical staining. The protective effect of ITLN1 on kidney function was evaluated by measuring changes in rat body weight and blood pressure, serum kidney injury markers, and kidney structure. The experimental results indicate that overexpression of ITLN1 can improve kidney function in rats with radiation-induced kidney injury by activating the Akt/GSK-3ß/Nrf2 signaling pathway, suppressing oxidative stress, cell apoptosis, inflammation, cellular senescence, and fibrosis. This study highlights the significant role of ITLN1 in regulating kidney injury, providing a novel target for future treatments of radiation-induced kidney injury.


Asunto(s)
Riñón , Animales , Ratas , Riñón/patología , Riñón/metabolismo , Riñón/efectos de la radiación , Masculino , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/etiología , Humanos , Traumatismos por Radiación/genética , Ratas Sprague-Dawley , Transducción de Señal , Traumatismos Experimentales por Radiación/metabolismo
3.
Int Immunopharmacol ; 132: 111945, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38555816

RESUMEN

BACKGROUND: Emodin, a natural anthraquinone derivative isolated from the roots of Rheum officinale Baill, has many pharmacological effects including anti-inflammatory, antioxidant, antiviral, antibacterial and anti-cancer. However, little is known about the effect of emodin on acute radiation proctitis (ARP). The present study was conducted to determine its effects and elucidate its mechanisms involving AKT/MAPK/NF-κB/VEGF pathways in ARP mice. METHODS: Total 60 C57BL/6 mice were divided randomly into control group, ARP group, AKT inhibitor MK-2206 group, and different doses of emodin groups. ARP mice were induced by 27 Gy of 6 MV X-ray pelvic local irradiation. MK-2206 was given orally for 2 weeks on alternate days. Emodin was administered daily by oral gavage for 2 weeks. Subsequently, all mice were sacrificed on day 15. The rectal tissues were obtained for further tests. The general signs score and the pathological grade were used to evaluate the severity of ARP. The expression of NF-κB, VEGF and AQP1 were determined by immunohistochemistry and western blot. The expression of p-AKT, p-ERK, p-JNK, p-p38, Bcl-2 and Bax were assessed using western blot. RESULTS: The worse general signs and damaged tissue structure of ARP mice were profoundly ameliorated by emodin. The expression of p-AKT, p-ERK, NF-κB, VEGF and AQP1 were significantly increased, resulting in the inflammation-induced angiogenesis in ARP mice. However, the expression of p-JNK and p-p38 were decreased, leading to the reduction of apoptosis in ARP mice. Excitedly, emodin reversed these changes, not only inhibited inflammation-induced angiogenesis, but also promoted apoptosis. Notably, the effects of emodin were similar to that of AKT inhibitor MK-2206, suggesting the involvement of AKT signaling in the effect of emodin. CONCLUSION: These results suggest that emodin attenuates ARP in mice, and the underlying mechanism might involve inhibition of the AKT/ERK/NF-κB/VEGF pathways and the induction of apoptosis mediated by JNK and p38.


Asunto(s)
Emodina , Ratones Endogámicos C57BL , FN-kappa B , Proctitis , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular , Animales , Emodina/farmacología , Emodina/uso terapéutico , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proctitis/tratamiento farmacológico , Proctitis/etiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ratones , Transducción de Señal/efectos de los fármacos , Traumatismos por Radiación/tratamiento farmacológico , Traumatismos por Radiación/patología , Masculino , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Compuestos Heterocíclicos con 3 Anillos/farmacología , Compuestos Heterocíclicos con 3 Anillos/uso terapéutico , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Traumatismos Experimentales por Radiación/patología , Traumatismos Experimentales por Radiación/metabolismo , Recto/patología , Recto/efectos de los fármacos
4.
Int J Radiat Biol ; 100(5): 767-776, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38442208

RESUMEN

PURPOSE: Toxicities from head and neck (H&N) radiotherapy (RT) may affect patient quality of life and can be dose-limiting. Proteins from the transforming growth factor beta (TGF-ß) family are key players in the fibrotic response. While TGF-ß1 is known to be pro-fibrotic, TGF-ß3 has mainly been considered anti-fibrotic. Moreover, TGF-ß3 has been shown to act protective against acute toxicities after radio- and chemotherapy. In the present study, we investigated the effect of TGF-ß3 treatment during fractionated H&N RT in a mouse model. MATERIALS AND METHODS: 30 C57BL/6J mice were assigned to three treatment groups. The RT + TGF-ß3 group received local fractionated H&N RT with 66 Gy over five days, combined with TGF-ß3-injections at 24-hour intervals. Animals in the RT reference group received identical RT without TGF-ß3 treatment. The non-irradiated control group was sham-irradiated according to the same RT schedule. In the follow-up period, body weight and symptoms of oral mucositis and lip dermatitis were monitored. Saliva was sampled at five time points. The experiment was terminated 105 d after the first RT fraction. Submandibular and sublingual glands were preserved, sectioned, and stained with Masson's trichrome to visualize collagen. RESULTS: A subset of mice in the RT + TGF-ß3 group displayed increased severity of oral mucositis and increased weight loss, resulting in a significant increase in mortality. Collagen content was significantly increased in the submandibular and sublingual glands for the surviving RT + TGF-ß3 mice, compared with non-irradiated controls. In the RT reference group, collagen content was significantly increased in the submandibular gland only. Both RT groups displayed lower saliva production after treatment compared to controls. TGF-ß3 treatment did not impact saliva production. CONCLUSIONS: When repeatedly administered during fractionated RT at the current dose, TGF-ß3 treatment increased acute H&N radiation toxicities and increased mortality. Furthermore, TGF-ß3 treatment may increase the severity of radiation-induced salivary gland fibrosis.


Asunto(s)
Fibrosis , Ratones Endogámicos C57BL , Glándulas Salivales , Estomatitis , Factor de Crecimiento Transformador beta3 , Animales , Factor de Crecimiento Transformador beta3/metabolismo , Ratones , Estomatitis/etiología , Estomatitis/patología , Glándulas Salivales/efectos de la radiación , Glándulas Salivales/patología , Modelos Animales de Enfermedad , Masculino , Traumatismos por Radiación/patología , Traumatismos por Radiación/etiología , Femenino , Traumatismos Experimentales por Radiación/patología
5.
Am J Pathol ; 194(6): 975-988, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38423356

RESUMEN

Radiation-induced enteritis, a significant concern in abdominal radiation therapy, is associated closely with gut microbiota dysbiosis. The mucus layer plays a pivotal role in preventing the translocation of commensal and pathogenic microbes. Although significant expression of REGγ in intestinal epithelial cells is well established, its role in modulating the mucus layer and gut microbiota remains unknown. The current study revealed notable changes in gut microorganisms and metabolites in irradiated mice lacking REGγ, as compared to wild-type mice. Concomitant with gut microbiota dysbiosis, REGγ deficiency facilitated the infiltration of neutrophils and macrophages, thereby exacerbating intestinal inflammation after irradiation. Furthermore, fluorescence in situ hybridization assays unveiled an augmented proximity of bacteria to intestinal epithelial cells in REGγ knockout mice after irradiation. Mechanistically, deficiency of REGγ led to diminished goblet cell populations and reduced expression of key goblet cell markers, Muc2 and Tff3, observed in both murine models, minigut organoid systems and human intestinal goblet cells, indicating the intrinsic role of REGγ within goblet cells. Interestingly, although administration of broad-spectrum antibiotics did not alter the goblet cell numbers or mucin 2 (MUC2) secretion, it effectively attenuated inflammation levels in the ileum of irradiated REGγ absent mice, bringing them down to the wild-type levels. Collectively, these findings highlight the contribution of REGγ in counteracting radiation-triggered microbial imbalances and cell-autonomous regulation of mucin secretion.


Asunto(s)
Enteritis , Microbioma Gastrointestinal , Células Caliciformes , Homeostasis , Ratones Noqueados , Animales , Enteritis/microbiología , Enteritis/metabolismo , Enteritis/patología , Ratones , Células Caliciformes/patología , Células Caliciformes/metabolismo , Humanos , Proteínas Asociadas a Pancreatitis/metabolismo , Mucina 2/metabolismo , Disbiosis/microbiología , Disbiosis/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Factor Trefoil-3/metabolismo , Ratones Endogámicos C57BL , Traumatismos por Radiación/metabolismo , Traumatismos por Radiación/microbiología , Traumatismos por Radiación/patología , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/patología , Traumatismos Experimentales por Radiación/microbiología
6.
Int J Radiat Biol ; 100(5): 689-708, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38306495

RESUMEN

PURPOSE: This research endeavor was undertaken to elucidate the impact of an innovative ascorbate formulation on the regeneration process of full-thickness excision wounds in a rat model exposed to whole-body gamma irradiation, replicating conditions akin to combat or radiation emergency scenarios. MATERIALS AND METHODS: We established a comprehensive rat model by optimizing whole body γ-radiation doses (5-9 Gy) and full-thickness excision wound sizes (1-3 cm2) to mimic radiation combined injury (RCI). The developed RCI model was used to explore the healing potential of ascorbate formulation. The study includes various treatment groups (i.e., sham control, radiation alone, wound alone, radiation + wound, and radiation + wound + formulation). The ascorbate formulation was applied twice daily, with a 12-hour gap between each application, starting 1 hour after the initiation of the wound. The healing potential of the formulation in the RCI context was evaluated over 14 days through hematological, molecular, and histological parameters. RESULTS: The combination of a 5 Gy radiation dose and a 1 cm2 wound was identified as the optimal setting to develop the RCI model for subsequent studies. The formulation was used topically immediately following RCI, and then twice daily until complete healing. Treatment with the ascorbate formulation yielded noteworthy outcomes and led to a substantial reduction (p < .05) in the wound area, accelerated epithelialization periods, and an increased wound contraction rate. The formulation's localized healing response improved organ weights, normalized blood parameters, and enhanced hematopoietic and immune systems. A gene expression study revealed the treatment up-regulated TGF-ß and FGF, and down-regulated PDGF-α, TNF-α, IL-1ß, IL-6, MIP-1α, and MCP-1 (p < .05). Histopathological assessments supported the formulation's effectiveness in restoring cellular architecture and promoting tissue regeneration. CONCLUSION: Topical application of the ascorbate formulation in RCI resulted in a significant improvement in delayed wound healing, leading to accelerated wound closure by mitigating the expression of inflammatory responses.


Asunto(s)
Administración Tópica , Ácido Ascórbico , Piel , Cicatrización de Heridas , Animales , Ácido Ascórbico/farmacología , Ácido Ascórbico/administración & dosificación , Ratas , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/efectos de la radiación , Piel/efectos de la radiación , Piel/efectos de los fármacos , Piel/lesiones , Piel/patología , Masculino , Modelos Animales de Enfermedad , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Traumatismos Experimentales por Radiación/patología , Ratas Sprague-Dawley , Rayos gamma , Irradiación Corporal Total
7.
Radiat Res ; 201(5): 406-417, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38319684

RESUMEN

The purpose of this investigation was to characterize the natural history of a murine total-abdominal-irradiation exposure model to measure gastrointestinal acute radiation injury. Male CD2F1 mice at 12 to 15 weeks old received total-abdominal irradiation using 4-MV linear accelerator X-rays doses of 0, 11, 13.5, 15, 15.75 and 16.5 Gy (2.75 Gy/min). Daily cage-side (i.e., in the animal housing room) observations of clinical signs and symptoms including body weights on all animals were measured up to 10 days after exposure. Jejunum tissues from cohorts of mice were collected at 1, 3, 7 and 10 days after exposure and radiation injury was assessed by histopathological analyses. Results showed time- and dose-dependent loss of body weight [for example at 7 days: 0.66 (±0.80) % loss for 0 Gy, 6.40 (±0.76) % loss at 11 Gy, 9.43 (±2.06) % loss at 13.5 Gy, 23.53 (± 1.91) % loss at 15 Gy, 29.97 (±1.16) % loss at 15.75 Gy, and 31.79 (±0.76) % loss at 16.5 Gy]. Negligible clinical signs and symptoms, except body weight changes, of radiation injury were observed up to 10 days after irradiation with doses of 11 to 15 Gy. Progressive increases in the severity of clinical signs and symptoms were found after irradiation with doses >15 Gy. Jejunum histology showed a progressive dose-dependent increase in injury. For example, at 7 days postirradiation, the percent of crypts, compared to controls, decreased to 82.3 (±9.5), 69.2 (±12.3), 45.4 (±11.9), 18.0 (±3.4), and 11.5 (± 1.8) with increases in doses from 11 to 16.5 Gy. A mucosal injury scoring system was used that mainly focused on changes in villus morphology damage (i.e., subepithelial spaces near the tips of the villi with capillary congestion, significant epithelial lifting along the length of the villi with a few denuded villus tips). Peak levels of total-abdominal irradiation induced effects on the mucosal injury score were seen 7 days after irradiation for doses ≥15 Gy, with a trend to show a decline after 7 days. A murine multiple-parameter gastrointestinal acute-radiation syndrome severity-scoring system was established based on clinical signs and symptoms that included measures of appearance (i.e., hunched and/or fluffed fur), respiratory rate, general (i.e., decreased mobility) and provoked behavior (i.e., subdued response to stimulation), weight loss, and feces/diarrhea score combined with jejunum mucosal-injury grade score. In summary, the natural-history radio-response for murine partial-body irradiation exposures is important for establishing a well-characterized radiation model system; here we established a multiple-parameter gastrointestinal acute-radiation syndrome severity-scoring system that provides a radiation injury gastrointestinal tissue-based assessment utility.


Asunto(s)
Síndrome de Radiación Aguda , Animales , Ratones , Masculino , Síndrome de Radiación Aguda/patología , Síndrome de Radiación Aguda/etiología , Relación Dosis-Respuesta en la Radiación , Yeyuno/efectos de la radiación , Yeyuno/patología , Modelos Animales de Enfermedad , Índice de Severidad de la Enfermedad , Tracto Gastrointestinal/efectos de la radiación , Tracto Gastrointestinal/patología , Peso Corporal/efectos de la radiación , Traumatismos Experimentales por Radiación/patología
8.
Radiat Res ; 201(5): 460-470, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38376474

RESUMEN

With the current volatile geopolitical climate, the threat of nuclear assault is high. Exposure to ionizing radiation from either nuclear incidents or radiological accidents often lead to major harmful consequences to human health. Depending on the absorbed dose, the symptoms of the acute radiation syndrome and delayed effects of acute radiation exposure (DEARE) can appear within hours, weeks to months. The lung is a relatively radiosensitive organ with manifestation of radiation pneumonitis as an acute effect, followed by apparent fibrosis in weeks or even months. A recently developed, first-of-its-kind murine model for partial-body irradiation (PBI) injury, which can be used to test potential countermeasures against multi-organ damage such as gastrointestinal (GI) tract and lungs was used for irradiation, with 2.5% bone marrow spared (BM2.5-PBI) from radiation exposure. Long-term damage to lungs from radiation was evaluated using µ-CT scans, pulmonary function testing, histopathological parameters and molecular biomarkers. Pulmonary fibrosis was detected by ground glass opacity observed in µ-CT scans of male and female C57BL/6J mice 6-7 months after BM2.5-PBI. Lung mechanics assessments pertaining to peripheral airways suggested fibrotic lungs with stiffer parenchymal lung tissue and reduced inspiratory capacity in irradiated animals 6-7 months after BM2.5-PBI. Histopathological evaluation of the irradiated lungs revealed presence of focal and diffuse pleural, and parenchymal inflammatory and fibrotic lesions. Fibrosis was confirmed by elevated levels of collagen when compared to lungs of age-matched naïve mice. These findings were validated by findings of elevated levels of pro-fibrotic biomarkers and reduction in anti-inflammatory proteins. In conclusion, a long-term model for radiation-induced pulmonary fibrosis was established, and countermeasures could be screened in this model for survival and protection/mitigation or recovery from radiation-induced pulmonary damage.


Asunto(s)
Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Fibrosis Pulmonar , Animales , Fibrosis Pulmonar/etiología , Fibrosis Pulmonar/patología , Ratones , Masculino , Femenino , Pulmón/efectos de la radiación , Pulmón/patología , Neumonitis por Radiación/patología , Neumonitis por Radiación/etiología , Traumatismos Experimentales por Radiación/patología , Traumatismos Experimentales por Radiación/etiología
9.
Stem Cell Res Ther ; 14(1): 325, 2023 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-37953266

RESUMEN

BACKGROUND: Stromal vascular fraction (SVF) treatment promoted the regeneration of the intestinal epithelium, limiting lethality in a mouse model of radiation-induced gastrointestinal syndrome (GIS). The SVF has a heterogeneous cell composition; the effects between SVF and the host intestinal immunity are still unknown. The specific role of the different cells contained in the SVF needs to be clarified. Monocytes-macrophages have a crucial role in repair and monocyte recruitment and activation are orchestrated by the chemokine receptors CX3CR1 and CCR2. METHODS: Mice exposed to abdominal radiation (18 Gy) received a single intravenous injection of SVF (2.5 × 106 cells), obtained by enzymatic digestion of inguinal fat tissue, on the day of irradiation. Intestinal immunity and regeneration were evaluated by flow cytometry, RT-PCR and histological analyses. RESULTS: Using flow cytometry, we showed that SVF treatment modulated intestinal monocyte differentiation at 7 days post-irradiation by very early increasing the CD11b+Ly6C+CCR2+ population in the intestine ileal mucosa and accelerating the phenotype modification to acquire CX3CR1 in order to finally restore the F4/80+CX3CR1+ macrophage population. In CX3CR1-depleted mice, SVF treatment fails to mature the Ly6C-MCHII+CX3CR1+ population, leading to a macrophage population deficit associated with proinflammatory environment maintenance and defective intestinal repair; this impaired SVF efficiency on survival. Consistent with a CD11b+ being involved in SVF-induced intestinal repair, we showed that SVF-depleted CD11b+ treatment impaired F4/80+CX3CR1+macrophage pool restoration and caused loss of anti-inflammatory properties, abrogating stem cell compartment repair and survival. CONCLUSIONS: These data showed that SVF treatment mitigates the GIS-involving immunomodulatory effect. Cooperation between the monocyte in SVF and the host monocyte defining the therapeutic properties of the SVF is necessary to guarantee the effective action of the SVF on the GIS.


Asunto(s)
Monocitos , Traumatismos Experimentales por Radiación , Fracción Vascular Estromal , Animales , Ratones , Tejido Adiposo , Intestinos , Macrófagos , Células del Estroma , Traumatismos Experimentales por Radiación/terapia
10.
Nan Fang Yi Ke Da Xue Xue Bao ; 43(10): 1815-1820, 2023 Oct 20.
Artículo en Chino | MEDLINE | ID: mdl-37933660

RESUMEN

OBJECTIVE: To observe the therapeutic effect of metformin on pathological pain in mice with radiation dermatitis and explore the underlying mechanism. METHODS: Thirty-two male adult ICR mice were randomized into normal control group, radiation dermatitis model group, metformin treatment (200 mg/kg) group and gabapentin (100 mg/kg) group (n=8).In the latter two groups, metformin treatment was administered after modeling via intraperitoneal injection and gabapentin by gavage on a daily basis for 16 days; the mice in the control group and model group received intraperitoneal injection of normal saline.After the last administration, radiation dermatitis was graded in each group.Mechanical withdraw threshold (MWT) and thermal withdrawal latency (TWL) of the mice were tested one day before and at 1, 4, 8, 12 and 16 days after modeling.Western blotting was used to measure the protein expression levels of p38MAPK, p-p38MAPK, NF-κB p65 and p-NF-κB p65 in the L4-L6 spinal cord, and the concentrations of IL-1ß, IL-6 and TNF-α in the spinal cord tissue were determined with ELISA. RESULTS: Compared with those in the control group, the mice in the other 3 groups showed obvious symptoms of radiation dermatitis after modeling (P<0.05), which were significantly alleviated by treatment with metformin (P<0.05).The mice in the model group exhibited significant decreases in MWT and TWL (P<0.05), which were improved by treatment with metformin and gabapentin (P<0.05).Compared with those in the model group, the levels of p-p38MAPK, p-NF-κB p65, IL-1ß, IL-6 and TNF-α in the spinal cord were significantly decreased in the mice after metformin treatment (P<0.05). CONCLUSION: Metformin can significantly ameliorate pathological pain symptoms in mice with radiation dermatitis possibly by inhibiting the activation of p38MAPK/NF-κB signaling pathway.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Metformina , FN-kappa B , Radiodermatitis , Animales , Masculino , Ratones , Gabapentina/uso terapéutico , Interleucina-6/metabolismo , Ratones Endogámicos ICR , FN-kappa B/metabolismo , Dolor , Factor de Necrosis Tumoral alfa/metabolismo , Metformina/uso terapéutico , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Radiodermatitis/tratamiento farmacológico
11.
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
12.
Expert Opin Drug Discov ; 18(7): 797-814, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37073409

RESUMEN

BACKGROUND: Animal models are vital for the development of radiation medical countermeasures for the prophylaxis or treatment of acute radiation syndrome and for the delayed effects of acute radiation exposure. Nonhuman primates (NHPs) play an important role in the regulatory approval of such agents by the United States Food and Drug Administration following the Animal Rule. Reliance on such animal models requires that such models are well characterized. METHODS: Data gathered from both male and female animals under the same conditions and gathered concurrently are limited; therefore, the authors compared and contrasted here the radiosensitivity of both male and female NHPs provided different levels of clinical support over a range of acute, total-body gamma irradiation, as well as the influence of age and body weight. RESULTS: Under matched experimental conditions, the authors observed only marginal, but clearly evident differences between acutely irradiated male and female NHPs relative to the measured response endpoints (rates of survival, blood cell changes, and cytokine fluctuations). These differences appeared to be accentuated by the level of exposure as well as by the nature of clinical support. CONCLUSION: Additional studies with both sexes under various experimental conditions and different radiation qualities run concurrently are needed.


Asunto(s)
Síndrome de Radiación Aguda , Traumatismos Experimentales por Radiación , Animales , Estados Unidos , Masculino , Femenino , Tolerancia a Radiación , Modelos Animales de Enfermedad , Síndrome de Radiación Aguda/tratamiento farmacológico , Macaca mulatta
13.
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
14.
Int J Mol Sci ; 24(6)2023 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-36982722

RESUMEN

Currently, there are no biomarkers to predict lethal lung injury by radiation. Since it is not ethical to irradiate humans, animal models must be used to identify biomarkers. Injury to the female WAG/RijCmcr rat has been well-characterized after exposure to eight doses of whole thorax irradiation: 0-, 5-, 10-, 11-, 12-, 13-, 14- and 15-Gy. End points such as SPECT imaging of the lung using molecular probes, measurement of circulating blood cells and specific miRNA have been shown to change after radiation. Our goal was to use these changes to predict lethal lung injury in the rat model, 2 weeks post-irradiation, before any symptoms manifest and after which a countermeasure can be given to enhance survival. SPECT imaging with 99mTc-MAA identified a decrease in perfusion in the lung after irradiation. A decrease in circulating white blood cells and an increase in five specific miRNAs in whole blood were also tested. Univariate analyses were then conducted on the combined dataset. The results indicated that a combination of percent change in lymphocytes and monocytes, as well as pulmonary perfusion volume could predict survival from radiation to the lungs with 88.5% accuracy (95% confidence intervals of 77.8, 95.3) with a p-value of < 0.0001 versus no information rate. This study is one of the first to report a set of minimally invasive endpoints to predict lethal radiation injury in female rats. Lung-specific injury can be visualized by 99mTc-MAA as early as 2 weeks after radiation.


Asunto(s)
Lesión Pulmonar , MicroARNs , Traumatismos Experimentales por Radiación , Traumatismos por Radiación , Humanos , Femenino , Ratas , Animales , Lesión Pulmonar/diagnóstico por imagen , Lesión Pulmonar/etiología , Pulmón/diagnóstico por imagen , Pulmón/efectos de la radiación , Tomografía Computarizada de Emisión de Fotón Único/métodos , MicroARNs/genética , Biomarcadores , Traumatismos Experimentales por Radiación/diagnóstico por imagen
15.
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
16.
Int J Mol Sci ; 24(4)2023 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-36835431

RESUMEN

Mitochondrion is an important organelle of eukaryotic cells and a critical target of ionizing radiation (IR) outside the nucleus. The biological significance and mechanism of the non-target effect originating from mitochondria have received much attention in the field of radiation biology and protection. In this study, we investigated the effect, role, and radioprotective significance of cytosolic mitochondrial DNA (mtDNA) and its associated cGAS signaling on hematopoietic injury induced by IR in vitro culture cells and in vivo total body irradiated mice in this study. The results demonstrated that γ-ray exposure increases the release of mtDNA into the cytosol to activate cGAS signaling pathway, and the voltage-dependent anion channel (VDAC) may contribute to IR-induced mtDNA release. VDAC1 inhibitor DIDS and cGAS synthetase inhibitor can alleviate bone marrow injury and ameliorate hematopoietic suppression induced by IR via protecting hematopoietic stem cells and adjusting subtype distribution of bone marrow cells, such as attenuating the increase of the F4/80+ macrophage proportion in bone marrow cells. The present study provides a new mechanistic explanation for the radiation non-target effect and an alternative technical strategy for the prevention and treatment of hematopoietic acute radiation syndrome.


Asunto(s)
Citosol , ADN Mitocondrial , Hematopoyesis , Mitocondrias , Nucleotidiltransferasas , Traumatismos Experimentales por Radiación , Animales , Ratones , Citosol/metabolismo , ADN Mitocondrial/metabolismo , Mitocondrias/metabolismo , Nucleotidiltransferasas/metabolismo , Transducción de Señal , Hematopoyesis/efectos de la radiación , Traumatismos Experimentales por Radiación/metabolismo
17.
Respir Res ; 24(1): 25, 2023 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-36694200

RESUMEN

BACKGROUND: Radiation-induced lung injury (RILI) is the most common and serious complication of chest radiotherapy. However, reported radioprotective agents usually lead to radiation resistance in tumor cells. The key to solving this problem is to distinguish between the response of tumor cells and normal lung epithelial cells to radiation damage. METHODS: RNA-Seq was used to recognize potential target of alleviating the progression of RILI as well as inhibiting tumor growth. The activation of NLRP3 inflammasome in lung epithelial cells was screened by qRT-PCR, western blotting, immunofluorescence, and ELISA. An in vivo model of RILI and in vitro conditioned culture model were constructed to evaluate the effect of NLRP3/interleukin-1ß on fibroblasts activation. ROS, ATP, and (NADP)+/NADP(H) level in lung epithelial cells was detected to explore the mechanism of NLRP3 inflammasome activation. The lung macrophages of the mice were deleted to evaluate the role of lung epithelial cells in RILI. Moreover, primary cells were extracted to validate the results obtained from cell lines. RESULTS: NLRP3 activation in epithelial cells after radiation depends on glycolysis-related reactive oxygen species accumulation. DPYSL4 is activated and acts as a negative regulator of this process. The NLRP3 inflammasome triggers interleukin-1ß secretion, which directly affects fibroblast activation, proliferation, and migration, eventually leading to lung fibrosis. CONCLUSIONS: Our study suggests that NLRP3 inflammasome activation in lung epithelial cells is essential for radiation-induced lung injury. These data strongly indicate that targeting NLRP3 may be effective in reducing radiation-induced lung injury in clinical settings.


Asunto(s)
Inflamasomas , Lesión Pulmonar , Traumatismos Experimentales por Radiación , Animales , Ratones , Células Epiteliales/metabolismo , Inflamasomas/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Pulmón/metabolismo , Lesión Pulmonar/etiología , Lesión Pulmonar/genética , Lesión Pulmonar/metabolismo , NADP/metabolismo , NADP/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Traumatismos Experimentales por Radiación/complicaciones , Traumatismos Experimentales por Radiación/genética , Traumatismos Experimentales por Radiación/metabolismo
18.
J Cell Mol Med ; 27(2): 246-258, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36579449

RESUMEN

Radiation-induced intestinal injury (RIII) is a common complication after radiation therapy in patients with pelvic, abdominal, or retroperitoneal tumours. Recently, in the model of DSS (Dextran Sulfate Sodium Salt) -induced intestinal inflammatory injury, it has been found in the study that transgenic mice expressing hVDR in IEC (Intestinal Epithelial Cell) manifest highly anti-injury properties in colitis, suggesting that activated VDR in the epithelial cells of intestine may inhibit colitis by protecting the mucosal epithelial barrier. In this study, we investigated the effect of the expression and regulation of VDR on the protection of RIII, and the radiosensitivity in vitro experiments, and explored the initial mechanism of VDR in regulating radiosensitivity of IEC. As a result, we found that the expression of VDR in intestinal tissues and cells in mice can be induced by ionizing radiation. VDR agonists are able to prolong the average survival time of mice after radiation and reduce the radiation-induced intestinal injury. For lack of vitamin D, the radiosensitivity of intestinal epithelial cells in mice increased, which can be reduced by VDR activation. Ensuing VDR activation, the radiation-induced intestinal stem cells damage is decreased, and the regeneration and differentiation of intestinal stem cells is promoted as well. Finally, on the basis of sequencing analysis, we validated and found that VDR may target the HIF/PDK1 pathway to mitigate RIII. We concluded that agonism or upregulation of VDR expression attenuates radiation-induced intestinal damage in mice and promotes the repair of epithelial damage in intestinal stem cells.


Asunto(s)
Colitis , Receptores de Calcitriol , Animales , Ratones , Colitis/patología , Sulfato de Dextran/efectos adversos , Células Epiteliales/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Traumatismos Experimentales por Radiación
19.
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
20.
Free Radic Res ; 56(5-6): 411-426, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36201846

RESUMEN

Oxidative stress injury and subsequent inflammatory response are considered to play critical roles in radiation-induced lung injury (RILI). Nuclear factor erythroid 2-related factor 2 (Nrf2) is a key transcription factor that regulates oxidative stress response and represses inflammation, but its therapeutic value in RILI remains elusive. Our previous studies have shown that the near-infrared (NIR) IR-61 dye evokes intracellular antioxidant defense by enhancing Nrf2 signaling and promoting anti-inflammatory effects. We established a model of RILI in mice exposed to whole-thoracic irradiation. The results showed that IR-61 treatment notably improved pulmonary functions by decreasing lung density and diminishing airway resistance. In addition, IR-61 significantly ameliorated radiation-induced inflammatory cell infiltration and proinflammatory cytokine (IL-1ß, IL-6, and TNF-α) release, thereby mitigating inflammatory response. Furthermore, IR-61 mitigated radiation-induced lung fibrosis by decreasing the collagen deposition and the levels of fibrogenesis-related factors (collagen I, collagen III, α-SMA, and fibronectin). More importantly, IR-61 was found to accumulate in the mitochondria of macrophages in irradiated lung tissues. Therefore, the functions of IR-61 in macrophages were further studied in irradiated macrophage cell lines, MH-s and RAW 264.7 in vitro. The results indicated that IR-61 upregulated the expression of Nrf2 and heme oxygenase-1 (HO-1) and decreased the levels of reactive oxygen species (ROS) and pro-inflammatory cytokines (IL-1ß and IL-6) in macrophages after radiation. In summary, our study suggests that IR-61 effectively mitigates RILI by activating Nrf2 signaling in irradiated lung tissues. In particular, Nrf2-mediated anti-inflammatory and antioxidant effects in irradiated lung tissue macrophages play critical roles in protecting against RILI.


Asunto(s)
Colorantes , Lesión Pulmonar , Factor 2 Relacionado con NF-E2 , Traumatismos Experimentales por Radiación , Animales , Ratones , Antioxidantes/metabolismo , Citocinas/metabolismo , Hemo-Oxigenasa 1/metabolismo , Interleucina-6 , Lipopolisacáridos , Pulmón , Lesión Pulmonar/tratamiento farmacológico , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/metabolismo , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Colorantes/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...