Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 890
Filtrar
1.
Gut Microbes ; 16(1): 2347722, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38706205

RESUMEN

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


Asunto(s)
Microbioma Gastrointestinal , Indoles , Ratones Endogámicos C57BL , Probióticos , Receptores de Hidrocarburo de Aril , Vía de Señalización Wnt , Animales , Ratones , Microbioma Gastrointestinal/efectos de los fármacos , Masculino , Humanos , Probióticos/administración & dosificación , Probióticos/farmacología , Receptores de Hidrocarburo de Aril/metabolismo , Indoles/metabolismo , Indoles/farmacología , Protectores contra Radiación/farmacología , Organoides/metabolismo , Traumatismos por Radiación/metabolismo , Traumatismos por Radiación/prevención & control , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/efectos de la radiación , Intestinos/microbiología , Intestinos/efectos de la radiación , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética
2.
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
3.
Mol Nutr Food Res ; 68(8): e2300745, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38581304

RESUMEN

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


Asunto(s)
Apoptosis , Flavanonas , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno , Canales Catiónicos TRPV , Animales , Flavanonas/farmacología , Humanos , Canales Catiónicos TRPV/metabolismo , Apoptosis/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Masculino , Ratones , Protectores contra Radiación/farmacología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/efectos de la radiación , Mucosa Intestinal/metabolismo , Células HCT116 , Canales de Calcio/metabolismo , Intestinos/efectos de los fármacos , Intestinos/efectos de la radiación , Calcio/metabolismo , Traumatismos por Radiación/tratamiento farmacológico
4.
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
5.
Curr Radiopharm ; 16(1): 57-63, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36056845

RESUMEN

AIM: In the current study, we aimed to mitigate radiation-induced small intestinal toxicity using post-irradiation treatment with nano-micelle curcumin. BACKGROUND: Small intestine is one of the most radiosensitive organs within the body. Wholebody exposure to an acute dose of ionizing radiation may lead to severe injuries to this tissue and may even cause death after some weeks. OBJECTIVE: This study aimed to evaluate histopathological changes in the small intestine following whole-body irradiation and treatment with nanocurcumin. MATERIALS AND METHODS: Forty male Nordic Medical Research Institute mice were grouped into control, treatment with 100 mg/kg nano-micelle curcumin, whole-body irradiation with cobalt-60 gamma-rays (dose rate of 60 cGy/min and a single dose of 7 Gy), and treatment with 100 mg/kg nano-micelle curcumin 1 day after whole-body irradiation for 4 weeks. Afterward, all mice were sacrificed for histopathological evaluation of their small intestinal tissues. RESULTS: Irradiation led to severe damage to villi, crypts, glands as well as vessels, leading to bleeding. Administration of nano-micelle curcumin after whole-body irradiation showed a statistically significant improvement in radiation toxicity of the duodenum, jejunum and ileum (including a reduction in infiltration of polymorphonuclear cells, villi length shortening, goblet cells injury, Lieberkühn glands injury and bleeding). Although treatment with nano-micelle curcumin showed increased bleeding in the ileum for non-irradiated mice, its administration after irradiation was able to reduce radiation-induced bleeding in the ileum. CONCLUSION: Treatment with nano-micelle curcumin may be useful for mitigation of radiationinduced gastrointestinal system toxicity via suppression of inflammatory cells' infiltration and protection against villi and crypt shortening.


Asunto(s)
Curcumina , Masculino , Ratones , Animales , Curcumina/farmacología , Radiofármacos , Intestino Delgado/patología , Intestino Delgado/efectos de la radiación , Íleon , Mucosa Intestinal/patología , Mucosa Intestinal/efectos de la radiación
6.
J Cell Mol Med ; 25(21): 10306-10312, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34595829

RESUMEN

Ionizing radiation (IR)-induced intestinal damage is the major and common injury of patients receiving radiotherapy. Urolithin A (UroA) is a metabolite of the intestinal flora of ellagitannin, a compound found in fruits and nuts such as pomegranates, strawberries and walnuts. UroA shows the immunomodulatory and anti-inflammatory capacity in various metabolic diseases. To evaluate the radioprotective effects, UroA(0.4, 2 and 10 mg/kg) were intraperitoneally injected to C57BL/6 male mice 48, 24, 1 h prior to and 24 h after 9.0Gy TBI. The results showed that UroA markedly upregulated the survival of irradiated mice, especially at concentration of 2 mg/kg. UroA improved the intestine morphology architecture and the regeneration ability of enterocytes in irradiated mice. Then, UroA significantly decreased the apoptosis of enterocytes induced by radiation. Additionally, 16S rRNA sequencing analysis showed the effect of UroA is associated with the recovery of the IR-induced intestinal microbacteria profile changes in mice. Therefore, our results determinated UroA could be developed as a potential candidate for radiomitigators in radiotherapy and accidental nuclear exposure. And the beneficial functions of UroA might be associated with the inhibition of p53-mediated apoptosis and remodelling of the gut microbes.


Asunto(s)
Cumarinas/farmacología , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/efectos de la radiación , Radiación Ionizante , Protectores contra Radiación/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Cumarinas/metabolismo , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/efectos de la radiación , Tracto Gastrointestinal/patología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Mucosa Intestinal/efectos de la radiación , Ratones , Dosis de Radiación
7.
Physiol Rep ; 9(15): e14960, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34337895

RESUMEN

Ionizing radiation causes dramatic change in the transport and barrier functions of the intestine. The degree of radiation damage rate depends primarily on the absorbed dose and post-irradiation time. Variety of experimental protocols providing different time points and doses exist, with the lack of a common approach. In this study, to develop a unified convenient experimental scheme, dose and time dependence of barrier and transport properties of rat jejunum following ionizing radiation exposure were examined. Male Wistar rats were exposed to total body X-ray irradiation (2, 5, or 10 Gy). The control group was subjected to sham irradiation procedure. Samples of rat jejunum were obtained at 24, 48, or 72 h post-irradiation. Transepithelial resistance, short circuit current (Isc ), and paracellular permeability for sodium fluorescein of jejunum samples were measured in an Ussing chamber; a histological examination was also performed. These parameters were significantly disturbed only 72 h after irradiation at a dose of 10 Gy, which was accompanied by loss of crypt and villi, inflammatory infiltrations, and disintegration of enterocytes. This suggests that found experimental point (72 h after 10 Gy exposure) is the most appropriate for future study using rat jejunum as a model.


Asunto(s)
Fluoresceína/metabolismo , Mucosa Intestinal/patología , Yeyuno/patología , Rayos X/efectos adversos , Animales , Relación Dosis-Respuesta en la Radiación , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efectos de la radiación , Yeyuno/metabolismo , Yeyuno/efectos de la radiación , Masculino , Permeabilidad , Ratas , Ratas Wistar , Factores de Tiempo
8.
Radiat Res ; 196(2): 204-212, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34043805

RESUMEN

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


Asunto(s)
Duodeno/efectos de la radiación , Íleon/efectos de la radiación , Intestino Delgado/efectos de la radiación , Yeyuno/efectos de la radiación , Animales , Duodeno/fisiopatología , Humanos , Íleon/fisiopatología , Mucosa Intestinal/fisiopatología , Mucosa Intestinal/efectos de la radiación , Intestino Delgado/fisiopatología , Intestinos/fisiopatología , Intestinos/efectos de la radiación , Yeyuno/fisiopatología , Macaca mulatta/fisiología , Primates/fisiología , Dosis de Radiación , Radiación Ionizante , Irradiación Corporal Total
9.
J Tradit Chin Med ; 41(2): 254-261, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33825405

RESUMEN

OBJECTIVE: To evaluate the efficacy of Liangxue Guyuan decoction ( LGD) on radiation-induced intestinal injury in rats, and the possible underlying mechanism of action. METHODS: A total of 255 male Sprague-Dawley rats were used. 15 rats were assigned to the control group and the remaining 240 rats were exposed to a 60Co source at a dose of 11 Gy. Irradiated rats were randomly divided into model, dexamethasone (DXM), low-dose LGD (LGDl), and high-dose LGD (LGDh) groups and treated for 11 d. The survival rate, weight of body, intestinal pathology and the expression of toll-like receptor 4 (TLR4), myeloid differentiation primary response 88 (MyD88), and nuclear factor-kappa B (NF-κB) were recorded. RESULTS: Radiation reduced the survival rate and weight of rats, destroyed the intestinal structure, induced an inflammatory reaction, and increased both protein and mRNA expression of TLR4, MyD88, and NF-κB in ileum. However, LGDh increased the survival rate, inhibited weight loss, alleviated inflammation and improve the expression of TLR4 pathway. CONCLUSION: LGD increased the survival rate and inhibit weight loss of irradiated rats, and reduced inflammation and intestinal injury. The underlying mechanism may involve regulation of the TLR4/MyD88/NF-κB pathway.


Asunto(s)
Medicamentos Herbarios Chinos/administración & dosificación , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/efectos de la radiación , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Traumatismos por Radiación/tratamiento farmacológico , Receptor Toll-Like 4/metabolismo , Animales , Humanos , Mucosa Intestinal/lesiones , Mucosa Intestinal/metabolismo , Masculino , Factor 88 de Diferenciación Mieloide/genética , FN-kappa B/genética , Traumatismos por Radiación/genética , Traumatismos por Radiación/metabolismo , Radiación Ionizante , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/genética
10.
Life Sci ; 278: 119546, 2021 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-33915129

RESUMEN

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


Asunto(s)
Ácidos Cumáricos/uso terapéutico , Intestinos/efectos de los fármacos , Intestinos/efectos de la radiación , Estrés Oxidativo/efectos de los fármacos , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Protectores contra Radiación/uso terapéutico , Animales , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Mucosa Intestinal/efectos de la radiación , Intestinos/patología , Masculino , Ratones Endogámicos C57BL , Piroptosis/efectos de los fármacos , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/patología , Radiación Ionizante
11.
Biochem Biophys Res Commun ; 554: 199-205, 2021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-33812084

RESUMEN

Radiation enteritis (RE) is the most common radiotherapy complication, and effective RE treatments are lacking. Resveratrol exerts beneficial effects on radiation injury. However, the effect of resveratrol in radiation-induced intestinal injury and the underlying mechanism remain unclear. Here, a C57BL/6 mouse model of RE was established and an intestinal epithelial cell line was used to evaluate the protective effects of resveratrol against radiation-induced intestinal injury and the underlying mechanisms. Resveratrol improved radiation-induced oxidative stress and cell apoptosis via upregulating antioxidant enzymes and downregulating p53 acetylation. In vivo, resveratrol-treated mice exhibited longer survival; longer villi; more intestinal crypt cells; upregulated expression of Ki67, catalase, and superoxide dismutase 2; and fewer inflammatory proteins and apoptotic cells. These protective effects were suppressed by inhibition of SIRT1. These results demonstrate that resveratrol can reduce radiation-induced intestinal injury by inhibiting oxidative stress and apoptosis via the SIRT1/FOXO3a and PI3K/AKT pathways.


Asunto(s)
Enteritis/prevención & control , Proteína Forkhead Box O3/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Traumatismos Experimentales por Radiación/prevención & control , Resveratrol/farmacología , Sirtuina 1/metabolismo , Animales , Antioxidantes/farmacología , Apoptosis , Línea Celular , Modelos Animales de Enfermedad , Enteritis/etiología , Enteritis/metabolismo , Enteritis/patología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Mucosa Intestinal/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo , Fosfatidilinositol 3-Quinasas/metabolismo , Traumatismos Experimentales por Radiación/etiología , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/patología , Radiación Ionizante , Ratas , Transducción de Señal
12.
Biosci Rep ; 41(3)2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33605406

RESUMEN

Acute radiation injury caused by high-dose radiation exposure severely impedes the application of radiotherapy in cancer management. To deeply understand the side effects of radiation on intestinal tract, an irradiation murine model was applied and evaluated. C57BL/6 mice were given 4 Gy non-myeloablative irradiation, 8 Gy myeloablative irradiation and non-irradiation (control), respectively. Results demonstrated that the 8 Gy myeloablative irradiations significantly damaged the gut barrier along with decreasing MECA32 and ZO-1. However, a slight increase in MECA32 and ZO-1 was detected in the 4 Gy non-myeloablative irradiations treatment from day 5 to day 10. Further, the irradiations affected the expression of P38 and JNK mitogen-activated protein kinase (MAPK) but not ERK1/2 MAPK signal pathway. Moreover, irradiation had adverse effects on hematopoietic system, altered the numbers and percentages of intestinal inflammatory cells. The IL-17/AhR had big increase in the gut of 4 Gy irradiation mice at day 10 compared with other groups. Both 8 Gy myeloablative and 4 Gy non-myeloablative irradiation disturbed the levels of short-chain fatty acids (SCFAs) in intestine. Meanwhile, high dosage of irradiation decreased the intestinal bacterial diversity and altered the community composition. Importantly, the fatty acids generating bacteria Bacteroidaceae and Ruminococcaceae played key roles in community distribution and SCFAs metabolism after irradiation. Collectively, the irradiation induced gut barrier damage with dosages dependent that led to the decreased p38 MAPK and increased JNK MAPK, unbalanced the mononuclear cells (MNCs) of gut, disturbed intestinal bacterial community and SCFAs level.


Asunto(s)
Mucosa Intestinal/efectos de la radiación , Traumatismos Experimentales por Radiación/metabolismo , Animales , Antígenos de Superficie/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Ácidos Grasos Volátiles/metabolismo , Microbioma Gastrointestinal , Interleucina-17/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , MAP Quinasa Quinasa 4/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Traumatismos Experimentales por Radiación/etiología , Traumatismos Experimentales por Radiación/patología , Receptores de Hidrocarburo de Aril/metabolismo , Irradiación Corporal Total/efectos adversos , Proteína de la Zonula Occludens-1/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
Cell Death Dis ; 12(2): 195, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33602915

RESUMEN

Amifostine has been the only small molecule radio-protector approved by FDA for decades; however, the serious adverse effects limit its clinical use. To address the toxicity issues and maintain the good potency, a series of modified small polycysteine peptides had been prepared. Among them, compound 5 exhibited the highest radio-protective efficacy, the same as amifostine, but much better safety profile. To confirm the correlation between the radiation-protective efficacy and the DNA binding capability, each of the enantiomers of the polycysteine peptides had been prepared. As a result, the L-configuration compounds had obviously higher efficacy than the corresponding D-configuration enantiomers; among them, compound 5 showed the highest DNA binding capability and radiation-protective efficacy. To our knowledge, this is the first study that has proved their correlations using direct comparison. Further exploration of the mechanism revealed that the ionizing radiation (IR) triggered ferroptosis inhibition by compound 5 could be one of the pathways for the protection effect, which was different from amifostine. In summary, the preliminary result showed that compound 5, a polycysteine as a new type of radio-protector, had been developed with good efficacy and safety profile. Further study of the compound for potential use is ongoing.


Asunto(s)
Ferroptosis/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Yeyuno/efectos de los fármacos , Pulmón/efectos de los fármacos , Péptidos/farmacología , Traumatismos por Radiación/prevención & control , Protectores contra Radiación/farmacología , Amifostina/farmacología , Animales , Línea Celular , ADN/metabolismo , Modelos Animales de Enfermedad , Ferroptosis/efectos de la radiación , Glutatión/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Células Madre Hematopoyéticas/efectos de la radiación , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Mucosa Intestinal/efectos de la radiación , Yeyuno/metabolismo , Yeyuno/patología , Yeyuno/efectos de la radiación , Peroxidación de Lípido/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Pulmón/efectos de la radiación , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Estrés Oxidativo/efectos de los fármacos , Péptidos/síntesis química , Péptidos/metabolismo , Dosis de Radiación , Traumatismos por Radiación/genética , Traumatismos por Radiación/metabolismo , Traumatismos por Radiación/patología , Protectores contra Radiación/síntesis química , Protectores contra Radiación/metabolismo , Ratas , Irradiación Corporal Total
14.
Cell Res ; 31(3): 259-271, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33420425

RESUMEN

The capacity of 3D organoids to mimic physiological tissue organization and functionality has provided an invaluable tool to model development and disease in vitro. However, conventional organoid cultures primarily represent the homeostasis of self-organizing stem cells and their derivatives. Here, we established a novel intestinal organoid culture system composed of 8 components, mainly including VPA, EPZ6438, LDN193189, and R-Spondin 1 conditioned medium, which mimics the gut epithelium regeneration that produces hyperplastic crypts following injury; therefore, these organoids were designated hyperplastic intestinal organoids (Hyper-organoids). Single-cell RNA sequencing identified different regenerative stem cell populations in our Hyper-organoids that shared molecular features with in vivo injury-responsive Lgr5+ stem cells or Clu+ revival stem cells. Further analysis revealed that VPA and EPZ6438 were indispensable for epigenome reprogramming and regeneration in Hyper-organoids, which functioned through epigenetically regulating YAP signaling. Furthermore, VPA and EPZ6438 synergistically promoted regenerative response in gut upon damage in vivo. In summary, our results demonstrated a new in vitro organoid model to study epithelial regeneration, highlighting the importance of epigenetic reprogramming that pioneers tissue repair.


Asunto(s)
Mucosa Intestinal/lesiones , Mucosa Intestinal/metabolismo , Organoides/lesiones , Organoides/metabolismo , Regeneración/efectos de los fármacos , Técnicas de Cultivo de Tejidos/métodos , Animales , Benzamidas/administración & dosificación , Compuestos de Bifenilo/administración & dosificación , Células Cultivadas , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis/metabolismo , Medios de Cultivo Condicionados/química , Sulfato de Dextran/efectos adversos , Modelos Animales de Enfermedad , Femenino , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/efectos de la radiación , Intestinos/lesiones , Intestinos/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Morfolinas/administración & dosificación , Organoides/efectos de los fármacos , Organoides/efectos de la radiación , Piridonas/administración & dosificación , Traumatismos por Radiación/tratamiento farmacológico , Traumatismos por Radiación/metabolismo , Transducción de Señal/genética , Células Madre/metabolismo , Resultado del Tratamiento , Ácido Valproico/administración & dosificación
15.
Cancer Lett ; 501: 20-30, 2021 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-33359449

RESUMEN

High-dose radiation exposure induces gastrointestinal (GI) stem cell death, resulting in denudation of the intestinal mucosa and lethality from GI syndrome, for which there is currently no effective therapy. Studying an intestinal organoid-based functional model, we found that Sirtuin1(SIRT1) inhibition through genetic knockout or pharmacologic inhibition significantly improved mouse and human intestinal organoid survival after irradiation. Remarkably, mice administered with two doseages of SIRT1 inhibitors at 24 and 96 h after lethal irradiation promoted Lgr5+ intestinal stem cell and crypt recovery, with improved mouse survival (88.89% of mice in the treated group vs. 0% of mice in the control group). Moreover, our data revealed that SIRT1 inhibition increased p53 acetylation, resulting in the stabilization of p53 and likely contributing to the survival of intestinal epithelial cells post-radiation. These results demonstrate that SIRT1 inhibitors are effective clinical countermeasures to mitigate GI toxicity from potentially lethal radiation exposure.


Asunto(s)
Enfermedades Gastrointestinales/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/farmacología , Intestinos/efectos de los fármacos , Niacinamida/farmacología , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Sirtuina 1/antagonistas & inhibidores , Acetilación , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Enfermedades Gastrointestinales/etiología , Enfermedades Gastrointestinales/metabolismo , Enfermedades Gastrointestinales/patología , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Mucosa Intestinal/efectos de la radiación , Intestinos/patología , Intestinos/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Organoides , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/patología , Sirtuina 1/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
16.
Phytomedicine ; 81: 153424, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33278782

RESUMEN

BACKGROUD: Exposure to high-dose radiation, such as after a nuclear accident or radiotherapy, elicits severe intestinal damage and is associated with a high mortality rate. In treating patients exhibiting radiation-induced intestinal dysfunction, countermeasures to radiation are required. In principle, the cellular event underlying radiation-induced gastrointestinal syndrome is intestinal stem cell (ISC) apoptosis in the crypts. High-dose irradiation induces the loss of ISCs and impairs intestinal barrier function, including epithelial regeneration and integrity. Notch signaling plays a critical role in the maintenance of the intestinal epithelium and regulates ISC self-renewal. Ghrelin, a hormone produced mainly by enteroendocrine cells in the gastrointestinal tract, has diverse physiological and biological functions. PURPOSE: We investigate whether ghrelin mitigates radiation-induced enteropathy, focusing on its role in maintaining epithelial function. METHODS: To investigate the effect of ghrelin in radiation-induced epithelial damage, we analyzed proliferation and Notch signaling in human intestinal epithelial cell. And we performed histological analysis, inflammatory response, barrier functional assays, and expression of notch related gene and epithelial stem cell using a mouse model of radiation-induced enteritis. RESULTS: In this study, we found that ghrelin treatment accelerated the reversal of radiation-induced epithelial damage including barrier dysfunction and defective self-renewing property of ISCs by activating Notch signaling. Exogenous injection of ghrelin also attenuated the severity of radiation-induced intestinal injury in a mouse model. CONCLUSION: These data suggest that ghrelin may be used as a potential therapeutic agent for radiation-induced enteropathy.


Asunto(s)
Ghrelina/farmacología , Enfermedades Intestinales/tratamiento farmacológico , Mucosa Intestinal/citología , Receptores Notch/metabolismo , Células Madre/efectos de la radiación , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Línea Celular , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Humanos , Enfermedades Intestinales/etiología , Enfermedades Intestinales/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efectos de la radiación , Masculino , Ratones Endogámicos C57BL , Traumatismos por Radiación , Protectores contra Radiación/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Células Madre/efectos de los fármacos , Células Madre/patología , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/efectos de la radiación
17.
Sci Rep ; 10(1): 22211, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33335275

RESUMEN

The only available option to treat radiation-induced hematopoietic syndrome is allogeneic hematopoietic cell transplantation, a therapy unavailable to many patients undergoing treatment for malignancy, which would also be infeasible in a radiological disaster. Stromal cells serve as critical components of the hematopoietic stem cell niche and are thought to protect hematopoietic cells under stress. Prior studies that have transplanted mesenchymal stromal cells (MSCs) without co-administration of a hematopoietic graft have shown underwhelming rescue of endogenous hematopoiesis and have delivered the cells within 24 h of radiation exposure. Herein, we examine the efficacy of a human bone marrow-derived MSC therapy delivered at 3 h or 30 h in ameliorating radiation-induced hematopoietic syndrome and show that pancytopenia persists despite MSC therapy. Animals exposed to radiation had poorer survival and experienced loss of leukocytes, platelets, and red blood cells. Importantly, mice that received a therapeutic dose of MSCs were significantly less likely to die but experienced equivalent collapse of the hematopoietic system. The cause of the improved survival was unclear, as complete blood counts, splenic and marrow cellularity, numbers and function of hematopoietic stem and progenitor cells, and frequency of niche cells were not significantly improved by MSC therapy. Moreover, human MSCs were not detected in the bone marrow. MSC therapy reduced crypt dropout in the small intestine and promoted elevated expression of growth factors with established roles in gut development and regeneration, including PDGF-A, IGFBP-3, IGFBP-2, and IGF-1. We conclude that MSC therapy improves survival not through overt hematopoietic rescue but by positive impact on other radiosensitive tissues, such as the intestinal mucosa. Collectively, these data reveal that MSCs could be an effective countermeasure in cancer patients and victims of nuclear accidents but that MSCs alone do not significantly accelerate or contribute to recovery of the blood system.


Asunto(s)
Hematopoyesis/efectos de la radiación , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Traumatismos por Radiación/mortalidad , Traumatismos por Radiación/terapia , Animales , Biopsia , Médula Ósea/metabolismo , Médula Ósea/patología , Médula Ósea/efectos de la radiación , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/efectos de la radiación , Modelos Animales de Enfermedad , Femenino , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/efectos de la radiación , Humanos , Inmunofenotipificación , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Mucosa Intestinal/efectos de la radiación , Masculino , Células Madre Mesenquimatosas/citología , Pancitopenia/etiología , Pancitopenia/metabolismo , Pancitopenia/patología , Pronóstico , Traumatismos por Radiación/patología , Radioterapia/efectos adversos , Resultado del Tratamiento
18.
Int J Oncol ; 57(6): 1307-1318, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33173975

RESUMEN

Enhancing the radioresponsiveness of colorectal cancer (CRC) is essential for local control and prognosis. However, consequent damage to surrounding healthy cells can lead to treatment failure. We hypothesized that short­chain fatty acids (SCFAs) could act as radiosensitizers for cancer cells, allowing the administration of a lower and safer dose of radiation. To test this hypothesis, the responses of three­dimensional­cultured organoids, derived from CRC patients, to radiotherapy, as well as the effects of combined radiotherapy with the SCFAs butyrate, propionate and acetate as candidate radiosensitizers, were evaluated via reverse transcription­quantitative polymerase chain reaction, immunohistochemistry and organoid viability assay. Of the three SCFAs tested, only butyrate suppressed the proliferation of the organoids. Moreover, butyrate significantly enhanced radiation­induced cell death and enhanced treatment effects compared with administration of radiation alone. The radiation­butyrate combination reduced the proportion of Ki­67 (proliferation marker)­positive cells and decreased the number of S phase cells via FOXO3A. Meanwhile, 3/8 CRC organoids were found to be non­responsive to butyrate with lower expression levels of FOXO3A compared with the responsive cases. Notably, butyrate did not increase radiation­induced cell death and improved regeneration capacity after irradiation in normal organoids. These results suggest that butyrate could enhance the efficacy of radiotherapy while protecting the normal mucosa, thus highlighting a potential strategy for minimizing the associated toxicity of radiotherapy.


Asunto(s)
Ácido Butírico/administración & dosificación , Quimioradioterapia Adyuvante/métodos , Neoplasias del Colon/terapia , Proteína Forkhead Box O3/metabolismo , Neoplasias del Recto/terapia , Anciano , Anciano de 80 o más Años , Técnicas de Cultivo de Célula , Colectomía , Colon/citología , Colon/efectos de los fármacos , Colon/patología , Colon/efectos de la radiación , Neoplasias del Colon/patología , Colonoscopía , Femenino , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Mucosa Intestinal/efectos de la radiación , Masculino , Persona de Mediana Edad , Organoides , Proctectomía , Neoplasias del Recto/patología , Recto/citología , Recto/efectos de los fármacos , Recto/patología , Recto/efectos de la radiación
19.
Sci Rep ; 10(1): 18300, 2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-33110120

RESUMEN

Gamma radiation is a commonly used adjuvant treatment for abdominally localized cancer. Since its therapeutic potential is limited due to gastrointestinal (GI) syndrome, elucidation of the regenerative response following radiation-induced gut injury is needed to develop a preventive treatment. Previously, we showed that Krüppel-like factor 4 (KLF4) activates certain quiescent intestinal stem cells (ISCs) marked by Bmi1-CreER to give rise to regenerating crypts following γ irradiation. In the current study, we showed that γ radiation-induced expression of p21Waf1/Cip1 in Bmi1-CreER cells is likely mitigated by MUSASHI-1 (MSI1) acting as a negative regulator of p21Waf1/Cip1 mRNA translation, which promotes exit of the Bmi1-CreER cells from a quiescent state. Additionally, Bmi1-specific Klf4 deletion resulted in decreased numbers of MSI1+ cells in regenerating crypts compared to those of control mice. We showed that KLF4 binds to the Msi1 promoter and activates its expression in vitro. Since MSI1 has been shown to be crucial for crypt regeneration, this finding elucidates a pro-proliferative role of KLF4 during the postirradiation regenerative response. Taken together, our data suggest that the interplay among p21Waf1/Cip1, MSI1 and KLF4 regulates Bmi1-CreER cell survival, exit from quiescence and regenerative potential upon γ radiation-induced injury.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Rayos gamma/efectos adversos , Mucosa Intestinal/efectos de la radiación , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas de Unión al ARN/metabolismo , Traumatismos Experimentales por Radiación/metabolismo , Células Madre/efectos de la radiación , Animales , Células HEK293 , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Factor 4 Similar a Kruppel , Ratones , Reacción en Cadena de la Polimerasa , Células Madre/metabolismo
20.
J Photochem Photobiol B ; 212: 112018, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32957067

RESUMEN

Inflammatory bowel diseases are debilitating illnesses characterized by severe inflammation of the gastrointestinal tract. Treatments currently available are expensive and ineffective. We here investigated the role of red-light emitting diode (LED) on dextran sodium sulfate (DSS)-induced colitis. DSS was added to the drinking water of male mice at days 0, 2, 4 and withdrawn at day 6. LED irradiation was performed daily for 90s from day 6 to 9 on the right and left sides of the ventral surface and beside the external anal region. LED treatment decreased the amount of crypt dysplasia/edema, inflammatory infiltrates and ulcers, attenuated apoptosis and increased proliferation of crypt cells. Also, LED treatment induced expression of annexin A1 in the damaged epithelium, preserved the organization of claudin-1 and skewed cytokine profiling towards a more anti-inflammatory status. Thus, LED treatment promotes structural protection and modulates the inflammatory response, constituting a potential non-invasive and low-cost combined therapy to help patients achieve disease remission.


Asunto(s)
Colitis/patología , Colitis/terapia , Sulfato de Dextran/farmacología , Fototerapia , Animales , Colitis/inducido químicamente , Mucosa Intestinal/patología , Mucosa Intestinal/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos C57BL , Semiconductores , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...