Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mol Cancer ; 23(1): 90, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38711083

RESUMEN

BACKGROUND: Metabolic reprogramming and epigenetic alterations contribute to the aggressiveness of pancreatic ductal adenocarcinoma (PDAC). Lactate-dependent histone modification is a new type of histone mark, which links glycolysis metabolite to the epigenetic process of lactylation. However, the role of histone lactylation in PDAC remains unclear. METHODS: The level of histone lactylation in PDAC was identified by western blot and immunohistochemistry, and its relationship with the overall survival was evaluated using a Kaplan-Meier survival plot. The participation of histone lactylation in the growth and progression of PDAC was confirmed through inhibition of histone lactylation by glycolysis inhibitors or lactate dehydrogenase A (LDHA) knockdown both in vitro and in vivo. The potential writers and erasers of histone lactylation in PDAC were identified by western blot and functional experiments. The potential target genes of H3K18 lactylation (H3K18la) were screened by CUT&Tag and RNA-seq analyses. The candidate target genes TTK protein kinase (TTK) and BUB1 mitotic checkpoint serine/threonine kinase B (BUB1B) were validated through ChIP-qPCR, RT-qPCR and western blot analyses. Next, the effects of these two genes in PDAC were confirmed by knockdown or overexpression. The interaction between TTK and LDHA was identified by Co-IP assay. RESULTS: Histone lactylation, especially H3K18la level was elevated in PDAC, and the high level of H3K18la was associated with poor prognosis. The suppression of glycolytic activity by different kinds of inhibitors or LDHA knockdown contributed to the anti-tumor effects of PDAC in vitro and in vivo. E1A binding protein p300 (P300) and histone deacetylase 2 were the potential writer and eraser of histone lactylation in PDAC cells, respectively. H3K18la was enriched at the promoters and activated the transcription of mitotic checkpoint regulators TTK and BUB1B. Interestingly, TTK and BUB1B could elevate the expression of P300 which in turn increased glycolysis. Moreover, TTK phosphorylated LDHA at tyrosine 239 (Y239) and activated LDHA, and subsequently upregulated lactate and H3K18la levels. CONCLUSIONS: The glycolysis-H3K18la-TTK/BUB1B positive feedback loop exacerbates dysfunction in PDAC. These findings delivered a new exploration and significant inter-relationship between lactate metabolic reprogramming and epigenetic regulation, which might pave the way toward novel lactylation treatment strategies in PDAC therapy.


Asunto(s)
Carcinoma Ductal Pancreático , Regulación Neoplásica de la Expresión Génica , Glucólisis , Histonas , L-Lactato Deshidrogenasa , Neoplasias Pancreáticas , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/genética , Humanos , Histonas/metabolismo , Animales , Línea Celular Tumoral , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/genética , Ratones , Retroalimentación Fisiológica , Epigénesis Genética , Carcinogénesis/metabolismo , Carcinogénesis/genética , Pronóstico , Proliferación Celular , Femenino
2.
Diabetologia ; 66(3): 535-550, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36331598

RESUMEN

AIMS/HYPOTHESIS: Glucagon receptor (GCGR) antagonism ameliorates hyperglycaemia and promotes beta cell regeneration in mouse models of type 2 diabetes. However, the underlying mechanisms remain unclear. The present study aimed to investigate the mechanism of beta cell regeneration induced by GCGR antagonism in mice. METHODS: The db/db mice and high-fat diet (HFD)+streptozotocin (STZ)-induced mice with type 2 diabetes were treated with antagonistic GCGR monoclonal antibody (mAb), and the metabolic variables and islet cell quantification were evaluated. Plasma cytokine array and liver RNA sequencing data were used to screen possible mediators, including fibroblast growth factor 21 (FGF21). ELISA, quantitative RT-PCR and western blot were applied to verify FGF21 change. Blockage of FGF21 signalling by FGF21-neutralising antibody (nAb) was used to clarify whether FGF21 was involved in the effects of GCGR mAb on the expression of beta cell identity-related genes under plasma-conditional culture and hepatocyte co-culture conditions. FGF21 nAb-treated db/db mice, systemic Fgf21-knockout (Fgf21-/-) diabetic mice and hepatocyte-specific Fgf21-knockout (Fgf21Hep-/-) diabetic mice were used to reveal the involvement of FGF21 in beta cell regeneration. A BrdU tracing study was used to analyse beta cell proliferation in diabetic mice treated with GCGR mAb. RESULTS: GCGR mAb treatment improved blood glucose control, and increased islet number (db/db 1.6±0.1 vs 0.8±0.1 per mm2, p<0.001; HFD+STZ 1.2±0.1 vs 0.5±0.1 per mm2, p<0.01) and area (db/db 2.5±0.2 vs 1.2±0.2%, p<0.001; HFD+STZ 1.0±0.1 vs 0.3±0.1%, p<0.01) in diabetic mice. The plasma cytokine array and liver RNA sequencing data showed that FGF21 levels in plasma and liver were upregulated by GCGR antagonism. The GCGR mAb induced upregulation of plasma FGF21 levels (db/db 661.5±40.0 vs 466.2±55.7 pg/ml, p<0.05; HFD+STZ 877.0±106.8 vs 445.5±54.0 pg/ml, p<0.05) and the liver levels of Fgf21 mRNA (db/db 3.2±0.5 vs 1.8±0.1, p<0.05; HFD+STZ 2.0±0.3 vs 1.0±0.2, p<0.05) and protein (db/db 2.0±0.2 vs 1.4±0.1, p<0.05; HFD+STZ 1.6±0.1 vs 1.0±0.1, p<0.01). Exposure to plasma or hepatocytes from the GCGR mAb-treated mice upregulated the mRNA levels of characteristic genes associated with beta cell identity in cultured mouse islets and a beta cell line, and blockage of FGF21 activity by an FGF21 nAb diminished this upregulation. Notably, the effects of increased beta cell number induced by GCGR mAb were attenuated in FGF21 nAb-treated db/db mice, Fgf21-/- diabetic mice and Fgf21Hep-/- diabetic mice. Moreover, GCGR mAb treatment enhanced beta cell proliferation in the two groups of diabetic mice, and this effect was weakened in Fgf21-/- and Fgf21Hep-/- mice. CONCLUSIONS/INTERPRETATION: Our findings demonstrate that liver-derived FGF21 is involved in the GCGR antagonism-induced beta cell regeneration in a mouse model of type 2 diabetes.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Células Secretoras de Glucagón , Ratones , Animales , Glucagón/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Glucagón/metabolismo , Diabetes Mellitus Experimental/metabolismo , Receptores de Glucagón/genética , Modelos Animales de Enfermedad , Hígado/metabolismo , Citocinas/metabolismo , Ratones Endogámicos C57BL
3.
Am J Physiol Endocrinol Metab ; 324(1): E97-E113, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36383639

RESUMEN

Glucagon-secreting pancreatic α-cells play pivotal roles in the development of diabetes. Glucagon promotes insulin secretion from ß-cells. However, the long-term effect of glucagon on the function and phenotype of ß-cells had remained elusive. In this study, we found that long-term glucagon intervention or glucagon intervention with the presence of palmitic acid downregulated ß-cell-specific markers and inhibited insulin secretion in cultured ß-cells. These results suggested that glucagon induced ß-cell dedifferentiation under pathological conditions. Glucagon blockage by a glucagon receptor (GCGR) monoclonal antibody (mAb) attenuated glucagon-induced ß-cell dedifferentiation. In primary islets, GCGR mAb treatment upregulated ß-cell-specific markers and increased insulin content, suggesting that blockage of endogenous glucagon-GCGR signaling inhibited ß-cell dedifferentiation. To investigate the possible mechanism, we found that glucagon decreased FoxO1 expression. FoxO1 inhibitor mimicked the effect of glucagon, whereas FoxO1 overexpression reversed the glucagon-induced ß-cell dedifferentiation. In db/db mice and ß-cell lineage-tracing diabetic mice, GCGR mAb lowered glucose level, upregulated plasma insulin level, increased ß-cell area, and inhibited ß-cell dedifferentiation. In aged ß-cell-specific FoxO1 knockout mice (with the blood glucose level elevated as a diabetic model), the glucose-lowering effect of GCGR mAb was attenuated and the plasma insulin level, ß-cell area, and ß-cell dedifferentiation were not affected by GCGR mAb. Our results proved that glucagon induced ß-cell dedifferentiation under pathological conditions, and the effect was partially mediated by FoxO1. Our study reveals a novel cross talk between α- and ß-cells and is helpful to understand the pathophysiology of diabetes and discover new targets for diabetes treatment.NEW & NOTEWORTHY Glucagon-secreting pancreatic α-cells can interact with ß-cells. However, the long-term effect of glucagon on the function and phenotype of ß-cells has remained elusive. Our new finding shows that long-term glucagon induces ß-cell dedifferentiation in cultured ß-cells. FoxO1 inhibitor mimicks whereas glucagon signaling blockage by GCGR mAb reverses the effect of glucagon. In type 2 diabetic mice, GCGR mAb increases ß-cell area, improves ß-cell function, and inhibits ß-cell dedifferentiation, and the effect is partially mediated by FoxO1.


Asunto(s)
Diabetes Mellitus Experimental , Insulinas , Ratones , Animales , Receptores de Glucagón/metabolismo , Glucagón/metabolismo , Glucemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Desdiferenciación Celular , Ratones Noqueados , Insulina/metabolismo , Proteína Forkhead Box O1
4.
Diabetes Metab Res Rev ; 39(3): e3607, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36565185

RESUMEN

AIMS: Sodium-glucose co-transporter 2 inhibitors, including dapagliflozin, improve ß cell function in type 2 diabetic individuals. Whether dapagliflozin can protect islet microvascular endothelial cells (IMECs) and thus contribute to the improvement of ß cell function remains unknown. MATERIALS AND METHODS: The db/db mice were treated with dapagliflozin or vehicle for 6 weeks. ß cell function, islet capillaries and the levels of inflammatory chemokines in IMECs were detected. The mouse IMEC cell line MS-1 cells were incubated with palmitate and/or dapagliflozin for 24 h. Angiogenesis and inflammatory chemokine levels were evaluated, and the involved signalling pathways were analysed. The mouse ß cell line MIN6 cells, in the presence or absence of co-culture with MS-1 cells, were treated with palmitate and/or dapagliflozin for 24 h. The expression of ß cell specific markers and insulin secretion in MIN6 cells were determined. RESULTS: Dapagliflozin significantly improved ß cell function, increased islet capillaries and decreased the levels of inflammatory chemokines of IMECs in db/db mice. In the palmitate-treated MS-1 cells, angiogenesis was enhanced and the levels of inflammatory chemokines were downregulated by dapagliflozin. Either a PI3K inhibitor or mTOR inhibitor eliminated the dapagliflozin-mediated effects. Importantly, dapagliflozin attenuated the palmitate-induced downregulation of ß cell function-related gene expression and insulin secretion in MIN6 cells co-cultured with MS-1 cells but not in those on mono-culture. CONCLUSIONS: Dapagliflozin restores islet vascularisation and attenuates the inflammation of IMECs in type 2 diabetic mice. The dapagliflozin-induced improvement of ß cell function is at least partially accounted for by its beneficial effects on IMECs in a PI3K/Akt-mTOR-dependent manner.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Islotes Pancreáticos , Enfermedades Vasculares , Ratones , Animales , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Células Endoteliales , Fosfatidilinositol 3-Quinasas/metabolismo , Islotes Pancreáticos/metabolismo , Compuestos de Bencidrilo/farmacología , Enfermedades Vasculares/metabolismo , Palmitatos/metabolismo
5.
Ecotoxicol Environ Saf ; 228: 113017, 2021 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-34823214

RESUMEN

Soil alkalization severely limits plant growth and development, however, the mechanisms of alkaline response in plants remain largely unknown. In this study, we performed physiological and transcriptomic analyses using two alfalfa cultivars (Medicago sativa L.) with different sensitivities to alkaline conditions. The chlorophyll content and shoot fresh mass drastically declined in the alkaline-sensitive cultivar Algonquin (AG) following alkaline treatment (0-25 mM Na2CO3 solution), while the alkaline-tolerant cultivar Gongnong NO.1 (GN) maintained relatively stable growth and chlorophyll content. Compared with AG, GN had higher contents of Ca2+ and Mg2+; the ratios of Ca2+ and Mg2+ to Na+, proline and soluble sugar, as well as higher enzyme activities of peroxidase (POD) and catalase (CAT) under the alkaline conditions. Furthermore, transcriptomic analysis identified three categories of alkaline-responsive differentially expressed genes (DEGs) between the two cultivars: 48 genes commonly induced in both the cultivars (CAR), 574 genes from the tolerant cultivar (TAR), and 493 genes from the sensitive cultivar (SAR). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses showed that CAR genes were mostly involved in phenylpropanoid biosynthesis, lipid metabolism, and DNA replication and repair; TAR genes were significantly enriched in metabolic pathways, such as biosynthesis of amino acids and secondary metabolites including flavonoids, and the MAPK signaling pathway; SAR genes were specifically enriched in vitamin B6 metabolism. Taken together, the results identified candidate pathways associated with genetic variation in response to alkaline stress, providing novel insights into the mechanisms underlying alkaline tolerance in alfalfa.

6.
Neurobiol Dis ; 132: 104588, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31470105

RESUMEN

Neuroinflammation may induce a phenotype switch to reactive astrogliosis in neurodegenerative disorders. The calcium-activated potassium channel (KCa3.1) is active in the phenotypic switch that occurs during astrogliosis in Alzheimer's disease and ischemic stroke. Here, transcriptome sequencing (RNA-Seq), immunohistochemistry, western blotting, pharmacological blockade, and calcium imaging were used to investigate astrocyte KCa3.1 activity in neuroinflammation, Tau accumulation, and insulin signaling deficits in male wild-type C57BL/6 and KCa3.1-/- knockout (KO) mice, and in primary astrocyte cultures. KCa3.1 deficiency in KO mice decreased lipopolysaccharide (LPS)-induced memory deficits, neuronal loss, glial activation, Tau phosphorylation, and insulin signaling deficits in vivo. KCa3.1 expression in astrocytes was associated with LPS-induced upregulation of the Orai1 store-operated Ca2+ channel protein. The KCa3.1 channel was found to regulate store-operated Ca2+ overload through an interaction with Orai1 in LPS-induced reactive astrocytes. The LPS-induced effects on KCa3.1 and Orai1 indirectly promoted astrogliosis-related changes via the PI3K/AKT/GSK3ß and NF-κB signaling pathways in vitro. Unbiased evaluation of RNA-Seq results for actively translated RNAs confirmed that substantial astrocyte diversity was associated with KCa3.1 deficiency. Our results suggest that KCa3.1 regulated astrogliosis-mediated neuroinflammation, Tau accumulation, and insulin signaling deficiency via PI3K/AKT/GSK3ß and NF-κB signaling pathways, and contributing to neuronal loss and memory deficits in this neuroinflammation mouse model.


Asunto(s)
Astrocitos/metabolismo , Gliosis/metabolismo , Inflamación/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Transducción de Señal/fisiología , Animales , Astrocitos/patología , Encéfalo/metabolismo , Encéfalo/patología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Inflamación/patología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
7.
Ecotoxicol Environ Saf ; 147: 897-904, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28968942

RESUMEN

Selenite (Se (IV)) and selenate (Se (IV)) have recently been demonstrated to be equally effective in inhibiting mercury (Hg) phytotoxicity to plants. This assertion is still unclear. In this study, we aimed to explore the potential effects of Se species (Se4+ and Se6+) on the inhibition of the mercury (Hg) bioavailability to pak choi in dry land. Pot experiments with exposure to different dosages of mercuric chloride (HgCl2) and selenite (Na2SeO3) or selenate (Na2SeO4) were treated. To compare the influence of Se (IV) and Se (VI) on the bioaccumulation and bioavailability of Hg, the levels of total Hg in different pak choi (Brassica chinensis L.) tissues (roots and shoots) and the distribution changes of Hg fractions in soil before planting and after harvest were determined as well as the Hg IR values in soils (relative binding intensity) were analyzed. Results showed that application Se (IV) reduced the concentrations of Hg in pak choi roots more than Se (VI). Hg concentrations were also decreased in pak choi shoots in Se (IV) treatments, while which notably increased in Se (VI) treatments. Thus, Se (IV) plays a more important role than Se (VI) in limiting the absorption and bioaccumulation of Hg in pak choi. Moreover, this inhibition may only significantly occur when Se (IV) is at an appropriate level (2.5mg/kg). In addition, the good correlations between the proportions of mobile Hg fractions (soluble and exchangeable fractions), IR values with the Hg concentrations in plants were observed. This affirmed the importance of the Hg fractions transformation and the IR indicator of Hg in the assessment of their bioavailability. Our findings regarding the importance of Se (IV) influence in reducing Hg bioaccumulation not only provided the correct appraisal about the effect of Se species on the inhibition of the Hg phytotoxicity to pak choi in dry land, but also be a good reference for selecting Se fertilizer forms (Se4+ or Se6+).


Asunto(s)
Brassica/efectos de los fármacos , Mercurio/toxicidad , Ácido Selénico/farmacología , Ácido Selenioso/farmacología , Contaminantes del Suelo/toxicidad , Suelo/química , Disponibilidad Biológica , Brassica/metabolismo , China , Fertilizantes , Mercurio/metabolismo , Raíces de Plantas/efectos de los fármacos , Raíces de Plantas/metabolismo , Contaminantes del Suelo/metabolismo
8.
Anal Chem ; 89(2): 1178-1184, 2017 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-27936613

RESUMEN

Nitrate (NO3-N), the main plant/microbial nitrogen source, has a fast turnover in soil driven by species transformation (nitrification/denitrification) and phyto/microbiota assimilation. The technique of diffusive gradients in thin films (DGT) is capable of a robust, low disturbance measurement of NO3-N but has not been implemented due to the absence of a binding layer suitable for deployment in soils. In this study, a new styrene divinylbenzene-based absorbent with amine functional groups (SIR-100-HP) was cast into an agarose gel support. The NO3-N ion selectivity of the SIR-100-HP/agarose binding layer was retained in the presence of high multivalent ion concentrations and was used successfully to acquire in situ NO3-N measurements in bulk soil. The kinetics of binding and the maximum binding capacity were determined. The total capacity of the DGT containing the SIR-100-HP/agarose binding phase was 667 µg of NO3-N. The performance of DGT was not affected by varying pH (3-8) or ionic strength (0-0.018 mol L-1), while anion competition effects at concentrations reflecting those in common agricultural soils were found to be negligible. Complete elution (100% efficiency) of NO3-N from the binding phase was achieved using a solution of 5% NaCl. This technique was validated in three contrasting soils. CDGT measurements were in excellent agreement with pore water NO3-N values. Two-dimensional NO3-N mapping of a profile of flooded rice paddy soil demonstrated the potential of this novel methodology for improved characterization of in situ N speciation for further understanding of bioavailability and biogeochemical processes of NO3-N in soils.


Asunto(s)
Resinas de Intercambio Aniónico/química , Nitratos/análisis , Suelo/química , Estireno/química , Compuestos de Vinilo/química , Adsorción , Agricultura , Difusión , Monitoreo del Ambiente/métodos , Cinética , Concentración Osmolar , Sefarosa/química
9.
J Neuroinflammation ; 14(1): 203, 2017 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-29037241

RESUMEN

BACKGROUND: Reactive astrogliosis is one of the significantly pathological features in ischemic stroke accompanied with changes in gene expression, morphology, and proliferation. KCa3.1 was involved in TGF-ß-induced astrogliosis in vitro and also contributed to astrogliosis-mediated neuroinflammation in neurodegeneration disease. METHODS: Wild type mice and KCa3.1-/- mice were subjected to permanent middle cerebral artery occlusion (pMCAO) to evaluate the infarct areas by 2,3,5-triphenyltetrazolium hydrochloride staining and neurological deficit. KCa3.1 channels expression and cell localization in the brain of pMCAO mice model were measured by immunoblotting and immunostaining. Glia activation and neuron loss was measured by immunostaining. DiBAC4 (3) and Fluo-4AM were used to measure membrane potential and cytosolic Ca2+ level in oxygen-glucose deprivation induced reactive astrocytes in vitro. RESULTS: Immunohistochemistry on pMCAO mice infarcts showed strong upregulation of KCa3.1 immunoreactivity in reactive astrogliosis. KCa3.1-/- mice exhibited significantly smaller infarct areas on pMCAO and improved neurological deficit. Both activated gliosis and neuronal loss were attenuated in KCa3.1-/- pMCAO mice. In the primary cultured astrocytes, the expressions of KCa3.1 and TRPV4 were increased associated with upregulation of astrogliosis marker GFAP induced by oxygen-glucose deprivation. The activation of KCa3.1 hyperpolarized membrane potential and, by promoting the driving force for calcium, induced calcium entry through TRPV4, a cation channel of the transient receptor potential family. Double-labeled staining showed that KCa3.1 and TRPV4 channels co-localized in astrocytes. Blockade of KCa3.1 or TRPV4 inhibited the phenotype switch of reactive astrogliosis. CONCLUSIONS: Our data suggested that KCa3.1 inhibition might represent a promising therapeutic strategy for ischemia stroke.


Asunto(s)
Isquemia Encefálica/metabolismo , Sistemas de Liberación de Medicamentos , Gliosis/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/deficiencia , Bloqueadores de los Canales de Potasio/administración & dosificación , Accidente Cerebrovascular/metabolismo , Animales , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/patología , Células Cultivadas , Sistemas de Liberación de Medicamentos/métodos , Gliosis/tratamiento farmacológico , Gliosis/patología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/patología
10.
Neurochem Res ; 42(10): 2712-2729, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28497343

RESUMEN

Previously we have demonstrated that brain-derived neurotrophic factor (BDNF) contributes to spinal long-term potentiation (LTP) and pain hypersensitivity through activation of GluN2B-containing N-methyl-D-aspartate (GluN2B-NMDA) receptors in rats following spinal nerve ligation (SNL). However, the molecular mechanisms by which BDNF impacts upon GluN2B-NMDA receptors and spinal LTP still remain unclear. In this study, we first documented that Fyn kinase-mediated phosphorylation of GluN2B subunit at tyrosine 1472 (pGluN2BY1472) was involved in BDNF-induced spinal LTP and pain hypersensitivity in intact rats. Second, we revealed a co-localization of Fyn and GluN2B-NMDA receptor in cultured dorsal horn neurons, implying that Fyn is a possible intermediate kinase linking BDNF/TrkB signaling with GluN2B-NMDA receptors in the spinal dorsal horn. Furthermore, we discovered that both SNL surgery and intrathecal active Fyn could induce an increased expression of dorsal horn pGluN2BY1472, as well as pain hypersensitivity in response to von Frey filaments stimuli; and more importantly, all these actions were effectively abrogated by pre-treatment with either PP2 or ifenprodil to respectively inhibit Fyn kinase and GluN2B-NMDA receptors activity. Moreover, we found that intrathecal administration of BDNF scavenger TrkB-Fc prior to SNL surgery, could prevent the nerve injury-induced increase of both pFynY420 and pGluN2BY1472 expression, and also inhibit the mechanical allodynia in neuropathic rats. Collectively, these results suggest that Fyn kinase-mediated pGluN2BY1472 is critical for BDNF-induced spinal LTP and pain hypersensitivity in SNL rats. Therefore, the BDNF-Fyn-GluN2B signaling cascade in the spinal dorsal horn may constitute a key mechanism underlying central sensitization and neuropathic pain development after peripheral nerve injury.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Potenciación a Largo Plazo/fisiología , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Hiperalgesia/metabolismo , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Neuralgia/metabolismo , Traumatismos de los Nervios Periféricos/metabolismo , Fosforilación , Ratas Sprague-Dawley , Nervios Espinales/metabolismo , Tirosina/metabolismo
11.
Zhongguo Yi Liao Qi Xie Za Zhi ; 38(5): 341-4, 2014 Sep.
Artículo en Zh | MEDLINE | ID: mdl-25597081

RESUMEN

According to the clinical requirements of cardiopulmonary bypass surgery, this paper established a simulation system for cardiac surgery which consists of venous reservoir, variable balance chamber, blood suction bag, ventricle suction bag, resistance valves, pressure gauges and tubings. Using the proposed system, perfusionists can mimic the implementation of pre-established surgery strategy, predict various abnormal conditions in the operation, and accordingly take the urgent actions so as to improve the success rate of surgery and to ensure the safety of patients.


Asunto(s)
Procedimientos Quirúrgicos Cardíacos/instrumentación , Puente Cardiopulmonar/instrumentación , Catéteres de Permanencia , Corazón , Humanos , Succión
12.
Diabetes ; 73(6): 926-940, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38471012

RESUMEN

Sodium-glucose cotransporter 2 inhibitors, efficacious antidiabetic agents that have cardiovascular and renal benefits, can promote pancreatic ß-cell regeneration in type 2 diabetic mice. However, the underlying mechanism remains unclear. In this study, we aimed to use multiomics to identify the mediators involved in ß-cell regeneration induced by dapagliflozin. We showed that dapagliflozin lowered blood glucose level, upregulated plasma insulin level, and increased islet area in db/db mice. Dapagliflozin reshaped gut microbiota and modulated microbiotic and plasmatic metabolites related to tryptophan metabolism, especially l-tryptophan, in the diabetic mice. Notably, l-tryptophan upregulated the mRNA level of glucagon-like peptide 1 (GLP-1) production-related gene (Gcg and Pcsk1) expression and promoted GLP-1 secretion in cultured mouse intestinal L cells, and it increased the supernatant insulin level in primary human islets, which was eliminated by GPR142 antagonist. Transplant of fecal microbiota from dapagliflozin-treated mice, supplementation of l-tryptophan, or treatment with dapagliflozin upregulated l-tryptophan, GLP-1, and insulin or C-peptide levels and promoted ß-cell regeneration in db/db mice. Addition of exendin 9-39, a GLP-1 receptor (GLP-1R) antagonist, or pancreatic Glp1r knockout diminished these beneficial effects. In summary, treatment with dapagliflozin in type 2 diabetic mice promotes ß-cell regeneration by upregulating GLP-1 production, which is mediated via gut microbiota and tryptophan metabolism.


Asunto(s)
Compuestos de Bencidrilo , Microbioma Gastrointestinal , Péptido 1 Similar al Glucagón , Glucósidos , Células Secretoras de Insulina , Regeneración , Triptófano , Animales , Compuestos de Bencidrilo/farmacología , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Péptido 1 Similar al Glucagón/metabolismo , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/fisiología , Triptófano/metabolismo , Ratones , Glucósidos/farmacología , Glucósidos/uso terapéutico , Regeneración/efectos de los fármacos , Humanos , Masculino , Insulina/metabolismo , Glucemia/metabolismo , Glucemia/efectos de los fármacos , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/microbiología , Ratones Endogámicos C57BL , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Diabetes Mellitus Experimental/metabolismo , Receptor del Péptido 1 Similar al Glucagón/metabolismo
13.
Asian J Androl ; 25(3): 331-338, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35848706

RESUMEN

Male diabetic individuals present a marked impairment in fertility; however, knowledge regarding the pathogenic mechanisms and therapeutic strategies is unsatisfactory. The new hypoglycemic drug dapagliflozin has shown certain benefits, such as decreasing the risk of cardiovascular and renal events in patients with diabetes. Even so, until now, the effects and underlying mechanisms of dapagliflozin on diabetic male infertility have awaited clarification. Here, we found that dapagliflozin lowered blood glucose levels, alleviated seminiferous tubule destruction, and increased sperm concentrations and motility in leptin receptor-deficient diabetic db/db mice. Moreover, the glucagon-like peptide-1 receptor (GLP-1R) antagonist exendin (9-39) had no effect on glucose levels but reversed the protective effects of dapagliflozin on testicular structure and sperm quality in db/db mice. We also found that dapagliflozin inhibited the testicular apoptotic process by upregulating the expression of the antiapoptotic protein B-cell lymphoma 2 (BCL2) and X-linked inhibitor of apoptosis protein (XIAP) and inhibiting oxidative stress by enhancing the antioxidant status, including total antioxidant capacity, total superoxide dismutase (SOD) activity, and glutathione peroxidase (GPx) activity, as well as decreasing the level of 4-hydroxynonenal (4-HNE). Exendin (9-39) administration partially reversed these effects. Furthermore, dapagliflozin upregulated the glucagon-like peptide-1 (GLP-1) level in plasma and GLP-1R expression by promoting AKT8 virus oncogene cellular homolog (Akt) phosphorylation in testicular tissue. Exendin (9-39) partially inhibited Akt phosphorylation. These results suggest that dapagliflozin protects against diabetes-induced spermatogenic dysfunction via activation of the GLP-1R/phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway. Our results indicate the potential effects of dapagliflozin against diabetes-induced spermatogenic dysfunction.


Asunto(s)
Diabetes Mellitus , Proteínas Proto-Oncogénicas c-akt , Ratones , Animales , Masculino , Proteínas Proto-Oncogénicas c-akt/metabolismo , Antioxidantes , Fosfatidilinositol 3-Quinasas/metabolismo , Semen/metabolismo
14.
Diabetes ; 72(5): 599-610, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36826938

RESUMEN

Dysfunction of glucagon-secreting α-cells participates in the progression of diabetes, and glucagon receptor (GCGR) antagonism is regarded as a novel strategy for diabetes therapy. GCGR antagonism upregulates glucagon and glucagon-like peptide 1 (GLP-1) secretion and, notably, promotes ß-cell regeneration in diabetic mice. Here, we aimed to clarify the role of GLP-1 receptor (GLP-1R) activated by glucagon and/or GLP-1 in the GCGR antagonism-induced ß-cell regeneration. We showed that in db/db mice and type 1 diabetic wild-type or Flox/cre mice, GCGR monoclonal antibody (mAb) improved glucose control, upregulated plasma insulin level, and increased ß-cell area. Notably, blockage of systemic or pancreatic GLP-1R signaling by exendin 9-39 (Ex9) or Glp1r knockout diminished the above effects of GCGR mAb. Furthermore, glucagon-neutralizing antibody (nAb), which prevents activation of GLP-1R by glucagon, also attenuated the GCGR mAb-induced insulinotropic effect and ß-cell regeneration. In cultured primary mouse islets isolated from normal mice and db/db mice, GCGR mAb action to increase insulin release and to upregulate ß-cell-specific marker expression was reduced by a glucagon nAb, by the GLP-1R antagonist Ex9, or by a pancreas-specific Glp1r knockout. These findings suggest that activation of GLP-1R by glucagon participates in ß-cell regeneration induced by GCGR antagonism in diabetic mice. ARTICLE HIGHLIGHTS: Glucagon receptor (GCGR) antagonism promotes ß-cell regeneration in type 1 and type 2 diabetic mice and in euglycemic nonhuman primates. Glucagon and glucagon-like peptide 1 (GLP-1) can activate the GLP-1 receptor (GLP-1R), and their levels are upregulated following GCGR antagonism. We investigated whether GLP-1R activated by glucagon and/or GLP-1 contributed to ß-cell regeneration induced by GCGR antagonism. We found that blockage of glucagon-GLP-1R signaling attenuated the GCGR monoclonal antibody-induced insulinotropic effect and ß-cell regeneration in diabetic mice. Our study reveals a novel mechanism of ß-cell regeneration and uncovers the communication between α-cells and ß-cells in regulating ß-cell mass.


Asunto(s)
Diabetes Mellitus Experimental , Células Secretoras de Glucagón , Ratones , Animales , Glucagón/metabolismo , Receptores de Glucagón/genética , Receptor del Péptido 1 Similar al Glucagón/genética , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Diabetes Mellitus Experimental/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Insulina/metabolismo , Células Secretoras de Glucagón/metabolismo , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/metabolismo , Regeneración
15.
iScience ; 25(7): 104567, 2022 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-35789836

RESUMEN

The deficiency of pancreatic ß-cells is the key pathogenesis of diabetes, while glucagon-secreting α-cells are another player in the development of diabetes. Here, we aimed to investigate the effects of glucagon receptor (GCGR) antagonism on ß-cell neogenesis in type 2 diabetic (T2D) mice and explore the origins of the neogenic ß-cells. We showed that GCGR monoclonal antibody (mAb) elevated plasma insulin level and increased ß-cell mass in T2D mice. By using α-cell lineage-tracing (glucagon -cre -ß-gal) mice and inducible Ngn3+ pancreatic endocrine progenitor lineage-tracing (Ngn3-CreERT2-tdTomato) mice, we found that GCGR mAb treatment promoted α-cell regression to progenitors, and induced Ngn3+ progenitor reactivation and differentiation toward ß-cells. Besides, GCGR mAb upregulated the expression levels of ß-cell regeneration-associated genes and promoted insulin secretion in primary mouse islets, indicative of a direct effect on ß-cell identity. Our findings suggest that GCGR antagonism not only increases insulin secretion but also promotes pro-α-cell-derived ß-cell neogenesis in T2D mice.

16.
Chronic Dis Transl Med ; 7(4): 276-286, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34786546

RESUMEN

BACKGROUND: The global prevalence of nonalcoholic fatty liver disease (NAFLD) is increasing. The pathogenesis of NAFLD is multifaceted, and the underlying mechanisms are elusive. We conducted data mining analysis to gain a better insight into the disease and to identify the hub genes associated with the progression of NAFLD. METHODS: The dataset GSE49541, containing the profile of 40 samples representing mild stages of NAFLD and 32 samples representing advanced stages of NAFLD, was acquired from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) were identified using the R programming language. The Database for Annotation, Visualization and Integrated Discovery (DAVID) online tool and Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database were used to perform the enrichment analysis and construct protein-protein interaction (PPI) networks, respectively. Subsequently, transcription factor networks and key modules were identified. The hub genes were validated in a mice model of high fat diet (HFD)-induced NAFLD and in cultured HepG2 cells by real-time quantitative PCR. RESULTS: Based on the GSE49541 dataset, 57 DEGs were selected and enriched in chemokine activity and cellular component, including the extracellular region. Twelve transcription factors associated with DEGs were indicated from PPI analysis. Upregulated expression of five hub genes (SOX9, CCL20, CXCL1, CD24, and CHST4), which were identified from the dataset, was also observed in the livers of HFD-induced NAFLD mice and in HepG2 cells exposed to palmitic acid or advanced glycation end products. CONCLUSION: The hub genes SOX9, CCL20, CXCL1, CD24, and CHST4 are involved in the aggravation of NAFLD. Our results offer new insights into the underlying mechanism of NAFLD progression.

17.
World J Stem Cells ; 13(1): 64-77, 2021 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-33584980

RESUMEN

Diabetes, one of the most common chronic diseases in the modern world, has pancreatic ß cell deficiency as a major part of its pathophysiological mechanism. Pancreatic regeneration is a potential therapeutic strategy for the recovery of ß cell loss. However, endocrine islets have limited regenerative capacity, especially in adult humans. Almost all hypoglycemic drugs can protect ß cells by inhibiting ß cell apoptosis and dedifferentiation via correction of hyperglycemia and amelioration of the consequent inflammation and oxidative stress. Several agents, including glucagon-like peptide-1 and γ-aminobutyric acid, have been shown to promote ß cell proliferation, which is considered the main source of the regenerated ß cells in adult rodents, but with less clarity in humans. Pancreatic progenitor cells might exist and be activated under particular circumstances. Artemisinins and γ-aminobutyric acid can induce α-to-ß cell conversion, although some disputes exist. Intestinal endocrine progenitors can transdeterminate into insulin-producing cells in the gut after FoxO1 deletion, and pharmacological research into FoxO1 inhibition is ongoing. Other cells, including pancreatic acinar cells, can transdifferentiate into ß cells, and clinical and preclinical strategies are currently underway. In this review, we summarize the clinical and preclinical agents used in different approaches for ß cell regeneration and make some suggestions regarding future perspectives for clinical application.

18.
J Diabetes Complications ; 35(2): 107797, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33293208

RESUMEN

AIMS: We aimed to investigate whether treatment with exenatide could ameliorate endothelial injury in patients with type 2 diabetes mellitus (T2DM), and to identify biomarkers for predicting amelioration of the endothelial injury induced by the treatment. METHODS: Ninety-three patients with T2DM were recruited and treated with exenatide for 16 weeks. Enzyme-linked immunosorbent assays were performed at baseline and after the treatment to measure serum levels of endothelial injury markers, including soluble thrombomodulin (sTM). Patients were categorized as responders (n = 47) or non-responders (n = 46) based on median changes in their sTM levels. Serum levels of metabolites at baseline were measured with non-targeted liquid chromatography-mass spectrometry. The results obtained were evaluated with multivariate analysis. RESULTS: Treatment with exenatide for 16 weeks resulted in reduced body weight and improved levels of fasting plasma glucose, 2-hour postprandial plasma glucose, and HbA1c in patients with T2DM (all P < 0.05). Compared with baseline, serum levels of endothelial injury markers including sTM were significantly lowered after the treatment. Metabolites presented at significantly different levels in responders versus non-responders were considered as biomarkers for a therapeutic response of sTM to the exenatide treatment. Among those identified, 4-hydroxyproline and 12-oxo-9(Z)-dodecenoic acid were found to correlate most closely with the exenatide-induced endothelial protection response. The specificity and sensitivity of the multi-metabolite signature model contained higher 4-hydroxyproline and lower 12-oxo-9(Z)-dodecenoic acid were 53.3% and 92.3%, respectively, and the area under receiver operating characteristic curve was 69.2% (P < 0.001). CONCLUSIONS: Treatment with exenatide for 16 weeks ameliorates endothelial injury in patients with T2DM. Endothelial protection benefit from exenatide treatment was effectively predicted by the specific metabolomic combination of higher 4-hydroxyproline and lower 12-oxo-9(Z)-dodecenoic acid.


Asunto(s)
Diabetes Mellitus Tipo 2 , Endotelio Vascular/fisiopatología , Exenatida , Hipoglucemiantes , Metabolómica , Biomarcadores , Glucemia , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Exenatida/uso terapéutico , Hemoglobina Glucada/análisis , Humanos , Hidroxiprolina , Hipoglucemiantes/uso terapéutico
19.
J Hazard Mater ; 383: 121196, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-31539663

RESUMEN

Lanthanum oxide nanoparticles (nano-La2O3) was used to develop a novel binding gel within an in situ passive sampler based on diffusive gradients in thin-films technique (NL-DGT) for measuring As(V), Sb(V), and V(V). Performance characteristics of NL-DGT were independent of pH (pH: 3.1-7.9 for As, 3.1-8.5 for V, and 3.1-6.5 for Sb) and ionic strength (0.1-500 mmol L-1 for As and V, and 0.1-200 mmol L-1 for Sb). No obvious competition effects among As, Sb, and V with different concentration ratios were found for NL-DGT measurement. Long term storage (8-188 d) of the nano-La2O3 gels in 0.01 mol L-1 NaNO3 at 4 °C did not affect their performance. During the field deployments in Yangtze and Jiuxiang River, NL-DGT measured concentrations of As and V were similar to those measured by the grab samples, while some differences were found for Sb between DGT and grab sampling because higher pH (∼8.0) in the studied rivers caused the performance deterioration of NL-DGT. Generally, the newly developed NL-DGT is suitable for monitoring As and V in freshwater from acidic to light alkaline and Sb in acidic and neutral water.

20.
Peptides ; 131: 170349, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32561493

RESUMEN

Glucagon is an essential regulator of glucose homeostasis, particularly in type 2 diabetes (T2D). Blocking the glucagon receptor (GCGR) in diabetic animals and humans has been shown to alleviate hyperglycemia and increase circulating glucagon-like peptide-1 (GLP-1) levels. However, the origin of the upregulated GLP-1 remains to be clarified. Here, we administered high-fat diet + streptozotocin-induced T2D mice and diabetic db/db mice with REMD 2.59, a fully competitive antagonistic human GCGR monoclonal antibody (mAb) for 12 weeks. GCGR mAb treatment decreased fasting blood glucose levels and increased plasma GLP-1 levels in the T2D mice. In addition, GCGR mAb upregulated preproglucagon gene expression and the contents of gut proglucagon-derived peptides, particularly GLP-1, in the small intestine and colon. Notably, T2D mice treated with GCGR mAb displayed a higher L-cell density in the small intestine and colon, which was associated with increased numbers of LK-cells coexpressing GLP-1 and glucose-dependent insulinotropic polypeptide and reduced L-cell apoptosis. Furthermore, GCGR mAb treatment upregulated GLP-1 production in the pancreas, which was detected at lower levels than in the intestine. Collectively, these results suggest that GCGR mAb can increase intestinal GLP-1 production and L-cell number by enhancing LK-cell expansion and inhibiting L-cell apoptosis in T2D.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/farmacología , Diabetes Mellitus Experimental/tratamiento farmacológico , Polipéptido Inhibidor Gástrico/genética , Péptido 1 Similar al Glucagón/genética , Receptores de Glucagón/genética , Animales , Apoptosis/genética , Glucemia/metabolismo , Colon/efectos de los fármacos , Colon/metabolismo , Diabetes Mellitus Experimental/etiología , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Dieta Alta en Grasa/efectos adversos , Ayuno/metabolismo , Polipéptido Inhibidor Gástrico/metabolismo , Regulación de la Expresión Génica , Péptido 1 Similar al Glucagón/metabolismo , Humanos , Intestino Delgado/efectos de los fármacos , Intestino Delgado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Proglucagón/genética , Proglucagón/metabolismo , Receptores de Glucagón/antagonistas & inhibidores , Receptores de Glucagón/metabolismo , Transducción de Señal , Estreptozocina/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA