Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 44(3): 357-365, 2022 Jun.
Artículo en Zh | MEDLINE | ID: mdl-35791930

RESUMEN

Objective To explore the effects of interleukin-6 (IL-6) gene knockout on the cognitive function and pathological changes in 5×FAD transgenic mice of Alzheimer's disease.Methods IL-6+/- mice were crossed with 5×FAD mice to establish the 5×FAD;IL-6-/- mouse model,and 3-month-old and 10-month-old mice were selected for experiments.The cognitive function of mice was detected by behavioral tests,and HE staining and ß-amyloid (Aß) immunohistochemical staining were performed to detect the pathological changes of mouse brain tissue.Results The number of 5×FAD;IL-6-/- model mice (3 months old,n=20;10 months old,n=5) and 5×FAD littermate control (3 months old,n=26;10 months old,n=24) conformed to the Mendel's law.Compared with that of the 5×FAD mice at the same age,the discrimination ratio of 3-month-old 5×FAD;IL-6-/- mice increased in the novel object recognition test (q=3.890,P=0.002).Morris water maze test results showed that the 3-month-old 5×FAD;IL-6-/- mice had longer time spent in target quadrant (q=3.797,P=0.012) and more times of crossing platform (q=2.505,P=0.017) than the 5×FAD mice at the same age.The results of immunohistochemical staining showed that IL-6 knockout reduced the Aß deposition in the hippocampus (q=13.490,P=0.002;q=45.680,P<0.001) and cortex (q=16.830,P=0.001;q=14.180,P=0.001) of 5×FAD mice.Conclusion IL-6 gene knockout can significantly improve the spatial memory and reduce the Aß deposition in the brain of 5×FAD mice.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Alzheimer/genética , Animales , Cognición , Modelos Animales de Enfermedad , Flavina-Adenina Dinucleótido , Técnicas de Inactivación de Genes , Interleucina-6 , Ratones , Ratones Noqueados
2.
Blood ; 123(20): 3105-15, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24622326

RESUMEN

Exposure to total body irradiation (TBI) induces not only acute hematopoietic radiation syndrome but also long-term or residual bone marrow (BM) injury. This residual BM injury is mainly attributed to permanent damage to hematopoietic stem cells (HSCs), including impaired self-renewal, decreased long-term repopulating capacity, and myeloid skewing. These HSC defects were associated with significant increases in production of reactive oxygen species (ROS), expression of p16(Ink4a) (p16) and Arf mRNA, and senescence-associated ß-galacotosidase (SA-ß-gal) activity, but not with telomere shortening or increased apoptosis, suggesting that TBI induces residual BM injury via induction of HSC premature senescence. This suggestion is supported by the finding that SA-ß-gal(+) HSC-enriched LSK cells showed more pronounced defects in clonogenic activity in vitro and long-term engraftment after transplantation than SA-ß-gal(-) LSK cells isolated from irradiated mice. However, genetic deletion of p16 and/or Arf had no effect on TBI-induced residual BM suppression and HSC senescence, because HSCs from irradiated p16 and/or Arf knockout (KO) mice exhibited changes similar to those seen in HSCs from wild-type mice after exposure to TBI. These findings provide important new insights into the mechanism by which TBI causes long-term BM suppression (eg, via induction of premature senescence of HSCs in a p16-Arf-independent manner).


Asunto(s)
Médula Ósea/patología , Médula Ósea/efectos de la radiación , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Células Madre Hematopoyéticas/patología , Células Madre Hematopoyéticas/efectos de la radiación , Animales , Médula Ósea/metabolismo , Células Cultivadas , Senescencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Técnicas de Inactivación de Genes , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Telómero/metabolismo , Telómero/patología , Telómero/efectos de la radiación , Irradiación Corporal Total
3.
Int J Mol Sci ; 17(6)2016 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-27338355

RESUMEN

Senescent hematopoietic stem cells (HSCs) accumulate with age and exposure to stress, such as total-body irradiation (TBI), which may cause long-term myelosuppression in the clinic. However, the methods available for long-term myelosuppression remain limited. Previous studies have demonstrated that sustained p38 mitogen-activated protein kinases (p38 MAPK) activation in HSCs following exposure to TBI in mice and the administration of its inhibitor twenty-four hours after TBI may partially prevent long-term myelosuppression. However, long-term myelosuppression is latent and identified long after the administration of radiation. In this study, we investigated the effects of SB203580 (a small molecule inhibitor of p38 MAPK) on long-term myelosuppression induced by TBI. Mice with hematopoietic injury were injected intraperitoneally with SB203580 every other day five times beginning 70 days after 6 Gy of (137)Cs γ ray TBI. Our results at 80 days demonstrated that SB203580 did not significantly improve the TBI-induced long-term reduction of peripheral blood cell and bone marrow nucleated cell (BMNC) counts, or defects in hematopoietic progenitor cells (HPCs) and HSC clonogenic function. SB203580 reduced reactive oxygen species (ROS) production and p-p38 expression; however, SB203580 had no effect on p16 expression in the HSCs of mice. In conclusion, these findings suggest that treatment with SB203580 70 days after TBI in mice inhibits the ROS-p38 oxidative stress pathway; however, it has no therapeutic effect on long-term myelosuppression induced by TBI.


Asunto(s)
Senescencia Celular/efectos de los fármacos , Senescencia Celular/efectos de la radiación , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Irradiación Corporal Total , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Células Sanguíneas/efectos de los fármacos , Células Sanguíneas/metabolismo , Células Sanguíneas/efectos de la radiación , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Ensayo de Unidades Formadoras de Colonias , Imidazoles/farmacología , Masculino , Ratones , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/efectos de la radiación , Piridinas/farmacología , Especies Reactivas de Oxígeno/metabolismo
4.
Int J Mol Sci ; 15(6): 10541-53, 2014 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-24927144

RESUMEN

Hematopoietic injury is the most common side effect of radiotherapy. However, the methods available for the mitigating of radiation injury remain limited. Xuebijing injection (XBJ) is a traditional Chinese medicine used to treat sepsis in the clinic. In this study, we investigated the effects of XBJ on the survival rate in mice with hematopoietic injury induced by γ ray ionizing radiation (IR). Mice were intraperitoneally injected with XBJ daily for seven days after total body irradiation (TBI). Our results showed that XBJ (0.4 mL/kg) significantly increased 30-day survival rates in mice exposed to 7.5 Gy TBI. This effect may be attributable to improved preservation of white blood cells (WBCs) and hematopoietic cells, given that bone marrow (BM) cells from XBJ-treated mice produced more granulocyte-macrophage colony forming units (CFU-GM) than that in the 2 Gy/TBI group. XBJ also decreased the levels of reactive oxygen species (ROS) by increasing glutathione (GSH) and superoxide dismutase (SOD) levels in serum and attenuated the increased BM cell apoptosis caused by 2 Gy/TBI. In conclusion, these findings suggest that XBJ enhances the survival rate of irradiated mice and attenuates the effects of radiation on hematopoietic injury by decreasing ROS production in BM cells, indicating that XBJ may be a promising therapeutic candidate for reducing hematopoietic radiation injury.


Asunto(s)
Apoptosis/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Rayos gamma , Células Madre Hematopoyéticas/efectos de los fármacos , Animales , Apoptosis/efectos de la radiación , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/efectos de la radiación , Ensayo de Unidades Formadoras de Colonias , Glutatión/sangre , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/efectos de la radiación , Masculino , Medicina Tradicional China , Ratones , Ratones Endogámicos ICR , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/sangre , Irradiación Corporal Total
5.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 36(5): 538-41, 2014 Oct.
Artículo en Zh | MEDLINE | ID: mdl-25360654

RESUMEN

Dipeptidyl peptidase-4 (DPP-4) is a protease that cleaves the peptides with alanine, praline, or other selective amino acids at the N-terminal penultimate position. The substrates of DPP-4 include many chemokines, colony-stimulating factors, and interleukins. Recent research has shown that DPP-4 can affect the hematopoietic stem and progenitor cells and transplantation by truncating the granulocyte colony stimulating factor. However, its regulatory effect on DPP-4 and most peptides truncation are still unknown. This review summarizes the recent advances in the DPP-4 research.


Asunto(s)
Dipeptidil Peptidasa 4/fisiología , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Humanos
6.
Eur J Haematol ; 91(3): 249-261, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23772810

RESUMEN

OBJECTIVES: Transfusional iron overload is of major concern in hematological disease. Iron-overload-related dyserythropoiesis and reactive oxygen species (ROS)-related damage to hematopoietic stem cell (HSC) function are major setbacks in treatment for such disorders. We therefore aim to investigate the effect of iron overload on hematopoiesis in the patients and explore the role of ROS in iron-induced oxidative damage in hematopoietic cells and microenvironment in vitro. PATIENTS AND METHODS: The hematopoietic colony-forming capacity and ROS level of bone marrow cells were tested before and after iron chelation therapy. In vitro, we first established an iron overload model of bone marrow mononuclear cells (BMMNC) and umbilical cord-derived mesenchymal stem cells (UC-MSC). ROS level, cell cycle, and apoptosis were measured by FACS. Function of cells was individually studied by Colony-forming cell (CFC) assay and co-culture system. Finally, ROS-related signaling pathway was also detected by Western blot. RESULTS: After administering deferoxamine (DFO), reduced blood transfusion, increased neutrophil, increased platelet, and improved pancytopenia were observed in 76.9%, 46.2%, 26.9%, and 15.4% of the patients, respectively. Furthermore, the colony-forming capacity of BMMNC from iron overload patient was deficient, and ROS level was higher, which were partially recovered following iron chelation therapy. In vitro, exposure of BMMNC to ferric ammonium citrate (FAC) for 24 h decreased the ratio of CD34(+) cell from 0.91 ± 0.12% to 0.39 ± 0.07%. Excessive iron could also induce apoptosis, arrest cell cycle, and decrease function of BMMNC and UC-MSC, which was accompanied by increased ROS level and stimulated p38MAPK, p53 signaling pathway. More importantly, N-acetyl-L-cysteine (NAC) or DFO could partially attenuate cell injury and inhibit the signaling pathway induced by excessive iron. CONCLUSIONS: Our study shows that iron overload injures the hematopoiesis by damaging hematopoietic cell and hematopoietic microenvironment, which is mediated by ROS-related signaling proteins.


Asunto(s)
Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Sobrecarga de Hierro/metabolismo , Hierro/metabolismo , Células Madre Mesenquimatosas/metabolismo , Estrés Oxidativo , Adulto , Anciano , Apoptosis , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Ciclo Celular , Proliferación Celular , Ensayo de Unidades Formadoras de Colonias , Deferoxamina/uso terapéutico , Femenino , Puntos de Control de la Fase G1 del Ciclo Celular , Células Madre Hematopoyéticas/citología , Humanos , Sobrecarga de Hierro/tratamiento farmacológico , Masculino , Persona de Mediana Edad , Transducción de Señal
7.
Biol Pharm Bull ; 36(6): 980-7, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23563593

RESUMEN

Peroxisome proliferator-activated receptor γ (PPARγ) is a unique target for insulin sensitizer agents. These drugs have been used for the clinical treatment of type 2 diabetes for almost twenty years. However, serious safety issues are associated with the PPARγ agonist thiazolidinediones (TZDs). Selective PPARγ modulators (SPPARMs) which retain insulin sensitization without TZDs-like side effects are emerging as a promising new generation of insulin sensitizers. C333H is a novel structure compound synthesized by our laboratory. In diabetic rodent models, C333H has insulin-sensitizing and glucose-lowering activity comparable to that of TZDs, and causes no significant increase in body weight or adipose tissue weight in db/db mice. In diabetic db/db mice, C333H elevated circulating high molecular weight adiponectin isoforms, decreased PPARγ 273 serine phosphorylation in brown adipose tissue and selectively modulated the expression of a subset of PPARγ target genes in adipose tissue. In vitro, C333H weakly recruited coactivator and weakly dissociated corepressor activity. These findings suggest that C333H has similar properties to SPPARMs and may be a potential therapeutic agent for the treatment of type 2 diabetes.


Asunto(s)
Tejido Adiposo Pardo/efectos de los fármacos , Diabetes Mellitus/metabolismo , Furanos/farmacología , Resistencia a la Insulina/fisiología , Oxazoles/farmacología , PPAR gamma/agonistas , Tejido Adiposo Pardo/crecimiento & desarrollo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Animales , Glucemia/análisis , Línea Celular Tumoral , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Insulina/sangre , Leptina/sangre , Masculino , Ratones , Obesidad/sangre , Obesidad/inducido químicamente , Tamaño de los Órganos/efectos de los fármacos , PPAR gamma/metabolismo , Ratas , Ratas Wistar , Glutamato de Sodio
8.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 35(5): 547-52, 2013 Oct.
Artículo en Zh | MEDLINE | ID: mdl-24183045

RESUMEN

OBJECTIVE: To establish a mouse model of iron overload by intraperitoneal injection of iron dextran and investigate the impact of iron overload on bone marrow hematopoiesis. METHODS: A total of 40 C57BL/6 mice were divided into control group, low-dose iron group (12.5 mg/ml), middle-dose iron group (25 mg/ml), and high-dose iron group (50 mg/ml). The control group received normal saline (0.2 ml), and the rest were injected with intraperitoneal iron dextran every three days for six weeks. Iron overload was confirmed by observing the bone marrow, hepatic, and splenic iron deposits and the bone marrow labile iron pool. In addition, peripheral blood and bone marrow mononuclear cells were counted and the hematopoietic function was assessed. RESULTS: Iron deposits in bone marrow, liver, and spleen were markedly increased in the mouse models. Bone marrow iron was deposited mostly within the matrix with no significant difference in expression of labile iron pool.Compared with control group, the ability of hematopoietic colony-forming in three interventional groups were decreased significantly (P<0.05). Bone marrow mononuclear cells counts showed no significant difference. The amounts of peripheral blood cells (white blood cells, red blood cells, platelets, and hemoglobin) in different iron groups showed no significant difference among these groups;although the platelets were decreased slightly in low-dose iron group [(780.7±39.60)×10(9)/L], middle dose iron group [(676.2±21.43)×10(9)/L], and high-dose iron group [(587.3±19.67)×10(9)/L] when compared with the control group [(926.0±28.23)×10(9)/L], there was no significant difference(P>0.05). CONCLUSIONS: The iron-overloaded mouse model was successfully established by intraperitoneal administration of iron dextran. Iron overload can damage the hepatic, splenic, and bone marrow hematopoietic function, although no significant difference was observed in peripheral blood count.


Asunto(s)
Médula Ósea/efectos de los fármacos , Modelos Animales de Enfermedad , Hematopoyesis/efectos de los fármacos , Sobrecarga de Hierro/fisiopatología , Complejo Hierro-Dextran/toxicidad , Animales , Médula Ósea/fisiopatología , Sobrecarga de Hierro/inducido químicamente , Complejo Hierro-Dextran/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Bazo/efectos de los fármacos
9.
Animal Model Exp Med ; 6(4): 329-336, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37642199

RESUMEN

The risk of internal and external exposure to ionizing radiation (IR) has increased alongside the development and implementation of nuclear technology. Therefore, serious security issues have emerged globally, and there has been an increase in the number of studies focusing on radiological prevention and medical countermeasures. Radioprotective drugs are particularly important components of emergency medical preparedness strategies for the clinical management of IR-induced injuries. However, a few drugs have been approved to date to treat such injuries, and the related mechanisms are not entirely understood. Thus, the aim of the present review was to provide a brief overview of the World Health Organization's updated list of essential medicines for 2023 for the proper management of national stockpiles and the treatment of radiological emergencies. This review also discusses the types of radiation-induced health injuries and the related mechanisms, as well as the development of various radioprotective agents, including Chinese herbal medicines, for which significant survival benefits have been demonstrated in animal models of acute radiation syndrome.


Asunto(s)
Síndrome de Radiación Aguda , Defensa Civil , Medicamentos Esenciales , Contramedidas Médicas , Protectores contra Radiación , Animales , Síndrome de Radiación Aguda/tratamiento farmacológico , Síndrome de Radiación Aguda/prevención & control , Radiación Ionizante , Protectores contra Radiación/farmacología , Protectores contra Radiación/uso terapéutico
10.
Animal Model Exp Med ; 6(2): 146-154, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37062934

RESUMEN

BACKGROUND: Busulfan (BU) is an alkylating agent used as a conditioning agent prior to hematopoietic stem cell (HSC) transplantation as it is known to be cytotoxic to host hematopoietic stem and progenitor cells. The susceptibility of HSCs to BU injury plays an important role in the myeloablative efficacy of BU. Different susceptibilities were demonstrated in genetically diverse (GD) mice in our preliminary research. METHODS: Three strains of GD mice with different susceptibilities to BU-induced HSC injury were used for screening biological markers of HSC injury susceptibility in urine. The urine proteins were analyzed using liquid chromatography coupled with tandem mass spectrometry to screen for differentially expressed proteins. Screening for possible biomarkers based on differences in protein expression abundance was validated using enzyme-linked immunoassay (ELISA). RESULTS: Functional analysis showed that the differential proteins were all involved in a series of biological pathways related to cellular senescence, apoptosis, and angiogenesis; whereas the differential proteins of the high-susceptible strain were enriched for the regulation of bone marrow microenvironment pathways, those of low-susceptible strain were enriched for the proapoptotic effect of GTPase pathways. Based on protein abundance differences, several urinary proteins that may be indicative of susceptibility were screened, and ELISA validation results showed that angiotensin-converting enzyme may be a potential biomarker predicting HSC susceptibility for BU conditioning. CONCLUSIONS: This study indicates that urinary protein levels can reflect differences in susceptibility to BU-induced HSC injury. Using GD mice to construct genetic difference models will provide preclinical data for screening BU-related biological markers.


Asunto(s)
Busulfano , Trasplante de Células Madre Hematopoyéticas , Ratones , Animales , Busulfano/farmacología , Células Madre Hematopoyéticas , Alquilantes/toxicidad , Espectrometría de Masas en Tándem/métodos
11.
Int Immunopharmacol ; 114: 109557, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36692947

RESUMEN

OBJECTIVE: Spermidine, a natural polyamine, possesses anti-oxidant, autophagy-regulation, and anti-aging properties. Elevated levels of oxidative stress, which was mediated the senescence of hematopoietic stem cells (HSCs) induced by radiation exposure, may further contribute to long-term myelosuppression. Therefore, this study investigated the protective effect of spermidine on the long-term damage of the hematopoietic system caused by radiation exposure. METHODS: In vitro experiments, bone marrow mononuclear cells (BMMNCs) of C57BL/6 mice were isolated and incubated with 5 mM spermidine for 30 min, then irradiated by 2 Gy X ray. The survival rate, proliferation, and differentiation ability of BMMNCs were detected. In vivo experiment, mice received 4 Gy total body irradiation (TBI), 3 mM spermidine were administered in the drinking water every day for 14 days prior to irradiation and then continued for 30 days after irradiation. Peripheral blood, bone marrow cell typing, level of reactive oxygen species (ROS), colony-forming ability of HSC, and transplantation-reconstitution capability were detected. RESULTS: In vitro experiments, spermidine significantly improved the survival rate of BMMNCs as well as the proliferation and differentiation ability of HSCs exposure to ionizing radiation (IR). In vivo, spermidine reduced levels of ROS in HSCs; spermidine attenuated long-term myeloid differentiation deviation induced by TBI. Spermidine promoted the proliferation and differentiation ability of stem cells, but failed to ameliorate the decreased engraftment capacity of bone marrow cells in mice exposed to TBI. CONCLUSION: This study demonstrated that spermidine could promote the recovery of IR-induced inhibition of proliferation and differentiation ability of HSCs, partly through antioxidant effects. Whether combining spermidine with other radioprotectants could further increase protective efficacy and reduce the long-term bone marrow injury needs further investigation.


Asunto(s)
Traumatismos por Radiación , Espermidina , Animales , Ratones , Especies Reactivas de Oxígeno , Espermidina/farmacología , Ratones Endogámicos C57BL , Células Madre Hematopoyéticas , Células de la Médula Ósea , Antioxidantes/farmacología , Irradiación Corporal Total
12.
Acta Pharmacol Sin ; 33(7): 888-96, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22705731

RESUMEN

AIM: To appraise the efficacy of Vam3 (Amurensis H), a dimeric derivative of resveratrol, at inhibiting cigarette smoke-induced autophagy. METHODS: Human bronchial epithelial cells were treated with cigarette smoke condensates, and a chronic obstructive pulmonary disease (COPD) model was established by exposing male BALB/c mice to cigarette smoke. The protein levels of the autophagic marker microtubule-associated protein 1A/1B-light chain 3 (LC3), Sirtuin 1 (Sirt1), and foxhead box O 3a (FoxO3a) were examined using Western blotting and Immunohistochemistry. LC3 punctae were detected by immunofluorescence. The levels of FoxO3a acetylation were examined by immunoprecipitation. The level of intracellular oxidation was assessed by detecting ROS and GSH-Px. RESULTS: Vam3 attenuated cigarette smoke condensate-induced autophagy in human bronchial epithelial cells, and restored the expression levels of Sirt1 and FoxO3a that had been reduced by cigarette smoke condensates. Similar protective effects of Vam3, reducing autophagy and restoring the levels of Sirt1 and FoxO3a, were observed in the COPD animal model. Additionally, Vam3 also diminished the oxidative stress that was induced by the cigarette smoke condensates. CONCLUSION: Vam3 decreases cigarette smoke-induced autophagy via up-regulating/restoring the levels of Sirt1 and FoxO3a and inhibiting the induced oxidative stress.


Asunto(s)
Antioxidantes/uso terapéutico , Autofagia/efectos de los fármacos , Bronquios/citología , Células Epiteliales/efectos de los fármacos , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Fumar/metabolismo , Estilbenos/uso terapéutico , Animales , Antioxidantes/química , Antioxidantes/farmacología , Línea Celular , Células Epiteliales/metabolismo , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Estrés Oxidativo/efectos de los fármacos , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/patología , Resveratrol , Sirtuina 1/genética , Sirtuina 1/metabolismo , Fumar/efectos adversos , Fumar/tratamiento farmacológico , Estilbenos/química , Estilbenos/farmacología
13.
Yao Xue Xue Bao ; 47(4): 417-20, 2012 Apr.
Artículo en Zh | MEDLINE | ID: mdl-22799020

RESUMEN

Sirtuin1 (Sirt1) is a NAD-dependent class III histone deacetylase (HDAC), and regulates pulmonary immune/inflammatory system and the aging process mainly through post-translational modification. Sirt1 could become a potential target for treatment of lung diseases due to participating in the development of a variety of lung diseases. In this paper, physiological characteristics, biological activities, modification regulations and its relationship with chronic obstructive pulmonary emphysema, asthma and lung cancer are reviewed.


Asunto(s)
Enfermedades Pulmonares/metabolismo , Sirtuina 1/metabolismo , Animales , Asma/metabolismo , Benzamidas/farmacología , Humanos , Neoplasias Pulmonares/metabolismo , Naftoles/farmacología , Procesamiento Proteico-Postraduccional , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Resveratrol , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/fisiología , Estilbenos/farmacología
14.
Yao Xue Xue Bao ; 47(6): 689-95, 2012 Jun.
Artículo en Zh | MEDLINE | ID: mdl-22919714

RESUMEN

Currently, about 300 million people worldwide are affected by asthma. Most of these sufferers inhale immunosuppressants (ie corticosteroids) and beta-adrenergic receptor agonists for their asthma treatment. However, about 5%-10% of patients of asthma have poor response to such treatment. Investigation of kinase signaling pathway and nuclear transcription factor as a target molecule in the treatment of allergic asthma has been the concern of scholars home and abroad. This paper reviewed inhibitors of kinase signaling pathway and nuclear transcription factors for the treatment of asthma.


Asunto(s)
Asma/enzimología , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Animales , Asma/tratamiento farmacológico , Humanos , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal
15.
Animal Model Exp Med ; 5(3): 288-296, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35656737

RESUMEN

Morphine is a widely used analgesic, but its use in clinical precision medicine is limited by the variance in response among individuals. Although previous studies have shown that individual differences in morphine can be explained in terms of pharmacodynamics and pharmacokinetics, genetic polymorphisms also play an important role. However, the genetic basis of different sensitivity and tolerance susceptibility to morphine remains ambiguous. Using 15 strains of inbred Genetic Diversity (GD) mice, a new resource with wide genetic and phenotypic variation, we demonstrated great variance in sensitivity to morphine analgesia and susceptibility to morphine tolerance between different GD strains. Among-individual variance in response to morphine analgesia in the population can be modeled in GD mice. Two loci respectively may be associated with the among-individual variance in morphine sensitivity and tolerance, confirming the role of genetic factors in among-individual different responses to morphine. These results indicate that GD mice may be a potential tool for the identification of new biomarkers to improve the clinical administration of morphine.


Asunto(s)
Analgesia , Morfina , Animales , Variación Genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Morfina/farmacología , Dolor
16.
Animal Model Exp Med ; 5(6): 565-574, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36376997

RESUMEN

BACKGROUND: Gastrointestinal (GI) injury is one of the most common side effects of radiotherapy. However, there is no ideal therapy method except for symptomatic treatment in the clinic. Xuebijing (XBJ) is a traditional Chinese medicine, used to treat sepsis by injection. In this study, the protective effects of XBJ on radiation-induced intestinal injury (RIII) and its mechanism were explored. METHODS: The effect of XBJ on survival of irradiated C57BL/6 mice was monitored. Histological changes including the number of crypts and the length of villi were evaluated by H&E. The expression of Lgr5+ intestinal stem cells (ISCs), Ki67+ cells, villin and lysozymes were examined by immunohistochemistry. The expression of cytokines in the intestinal crypt was detected by RT-PCR. DNA damage and apoptosis rates in the small intestine were also evaluated by immunofluorescence. RESULTS: In the present study, XBJ improved the survival rate of the mice after 8.0 and 9.0 Gy total body irradiation (TBI). XBJ attenuated structural damage of the small intestine, maintained regenerative ability and promoted proliferation and differentiation of crypt cells, decreased apoptosis rate and reduced DNA damage in the intestine. Elevation of IL-6 and TNF-α was limited, but IL-1, TNF-𝛽 and IL-10 levels were increased in XBJ-treated group after irradiation. The expression of Bax and p53 were decreased after XBJ treatment. CONCLUSIONS: Taken together, XBJ provides a protective effect on RIII by inhibiting inflammation and blocking p53-related apoptosis pathway.


Asunto(s)
Medicamentos Herbarios Chinos , Proteína p53 Supresora de Tumor , Ratones , Animales , Proteína p53 Supresora de Tumor/metabolismo , Ratones Endogámicos C57BL , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Citocinas/metabolismo
17.
Yao Xue Xue Bao ; 46(4): 395-9, 2011 Apr.
Artículo en Zh | MEDLINE | ID: mdl-21751492

RESUMEN

This study is to investigate the protective effects of the SB203580 against radiation induced mortality and intestinal injury of mice. A total of 67 male C57BL/6 mice (20.0-22.0 g) were matched according to body weight and randomly assigned to one of three groups: control, total body irradiation exposure (IR, 7.2 Gy) only, and IR (7.2 Gy) + SB203580 (15 mg x kg(-1)). 30 days survival rate was observed in the experiment. In intestinal injury experiment, the expression levels of caspase-3, Ki67, p53 and p-p38 were assayed in the mice intestine crypts. The results showed that the 30 days survival rate was 100% (control), 0 (IR) and 40% (IR+ SB203580), separately. Compared to the IR groups, the positive cells of caspase-3, p53 and p-p38 in crypt cells decreased 33.00%, 21.78% and 34.63%, respectively. The rate of positive cells of Ki67 increased 37.96%. Significant difference was found between all of them (P < 0.01). SB203580 potently protected against radiation-induced lethal and intestinal injury in mice, and it may be a potential radio protector.


Asunto(s)
Apoptosis/efectos de la radiación , Imidazoles/farmacología , Intestinos/efectos de los fármacos , Piridinas/farmacología , Traumatismos Experimentales por Radiación , Protectores contra Radiación/farmacología , Animales , Caspasa 3/metabolismo , Inhibidores Enzimáticos/farmacología , Mucosa Intestinal/metabolismo , Intestinos/patología , Antígeno Ki-67/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/mortalidad , Traumatismos Experimentales por Radiación/patología , Distribución Aleatoria , Proteína p53 Supresora de Tumor/metabolismo , Irradiación Corporal Total , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
Cell Death Dis ; 12(6): 527, 2021 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-34023858

RESUMEN

Radiation-induced pulmonary fibrosis (RIPF) is a late toxicity of therapeutic radiation in clinic with poor prognosis and limited therapeutic options. Previous results have shown that senescent cells, such as fibroblast and type II airway epithelial cell, are strongly implicated in pathology of RIPF. However, the role of senescent macrophages in the development RIPF is still unknown. In this study, we report that ionizing radiation (IR) increase cellular senescence with higher expression of senescence-associated ß-galactosidase (SA-ß-Gal) and senescence-specific genes (p16, p21, Bcl-2, and Bcl-xl) in irradiated bone marrow-derived monocytes/macrophages (BMMs). Besides, there's a significant increase in the expression of pro-fibrogenic factors (TGF-ß1 and Arg-1), senescence-associated secretory phenotype (SASP) proinflammatory factors (Il-1α, Il-6, and Tnf-α), SASP chemokines (Ccl2, Cxcl10, and Ccl17), and SASP matrix metalloproteinases (Mmp2, Mmp9 and Mmp12) in BMMs exposed to 10 Gy IR. In addition, the percentages of SA-ß-Gal+ senescent macrophages are significantly increased in the macrophages of murine irradiated lung tissue. Moreover, robustly elevated expression of p16, SASP chemokines (Ccl2, Cxcl10, and Ccl17) and SASP matrix metalloproteinases (Mmp2, Mmp9, and Mmp12) is observed in the macrophages of irradiated lung, which might stimulate a fibrotic phenotype in pulmonary fibroblasts. In summary, irradiation can induce macrophage senescence, and increase the secretion of SASP in senescent macrophages. Our findings provide important evidence that senescent macrophages might be the target for prevention and treatment of RIPF.


Asunto(s)
Senescencia Celular/fisiología , Macrófagos/fisiología , Neumonitis por Radiación/patología , Animales , Células Cultivadas , Senescencia Celular/efectos de la radiación , Quimiocinas/metabolismo , Citocinas/metabolismo , Pulmón/metabolismo , Pulmón/patología , Pulmón/efectos de la radiación , Macrófagos/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos C57BL , Fibrosis Pulmonar/etiología , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/patología , Neumonitis por Radiación/metabolismo , Radiación Ionizante
19.
Oxid Med Cell Longev ; 2020: 8308616, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32774687

RESUMEN

Sitagliptin, an inhibitor of the dipeptidyl peptidase IV (DPP4), has been implicated in the regulation of type 2 diabetes. However, the role and mechanism of sitagliptin administration in total body irradiation (TBI)- induced hematopoietic cells injury are unclear. In this study, we demonstrated that sitagliptin had therapeutic effects on hematopoietic damage, which protected mice from 7.5 Gy TBI-induced death, increased the numbers and colony formation ability of hematopoietic cells. These therapeutic effects might be attributed to the inhibition of NOX4-mediated oxidative stress in hematopoietic cells, and the alleviation of inflammation was also helpful. Therefore, sitagliptin has potential as an effective radiotherapeutic agent for ameliorating TBI-induced hematopoietic injury.


Asunto(s)
Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Neoplasias Hematológicas/tratamiento farmacológico , Fosfato de Sitagliptina/uso terapéutico , Irradiación Corporal Total/métodos , Animales , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Neoplasias Hematológicas/mortalidad , Masculino , Ratones , Fosfato de Sitagliptina/farmacología , Análisis de Supervivencia
20.
Biochem Biophys Res Commun ; 379(4): 1107-13, 2009 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-19161979

RESUMEN

Gain of chromosome 1q is a common event in many kinds of carcinomas. The Cks1 gene, located at 1q21, is required for p27 ubiquitination by the SCF(skp2) ubiquitinating machinery. In the present study, we found that Cks1 gene amplification was highly correlated with protein overexpression. Statistical analysis showed that amplification and overexpression of Cks1 were strongly associated with lymph node metastasis and poor prognosis. At the molecular level, knockdown of Cks1 expression by RNA interference inhibited the growth of MDA-MB-231 cells, damaged cell migration and invasion ability. Knockdown of Cks1 expression promoted apoptosis of breast cancer cells and a wobble mutant of Cks1 that was resistant to Cks1 siRNA can rescue this effect. Overexpression of Cks1 inhibited the apoptosis of breast cancer cells through the MEK-Erk pathway. These data suggest that Cks1 is an oncogene in the 1q21 amplicon and plays an important role for breast cancer development.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteínas Portadoras/biosíntesis , Quinasas Ciclina-Dependientes/biosíntesis , Amplificación de Genes , Regulación Neoplásica de la Expresión Génica , Oncogenes , Apoptosis/genética , Neoplasias de la Mama/enzimología , Quinasas CDC2-CDC28 , Proteínas Portadoras/genética , Movimiento Celular/genética , Cromosomas Humanos Par 1/genética , Quinasas Ciclina-Dependientes/genética , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Invasividad Neoplásica , ARN Interferente Pequeño/genética , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA