Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Phytomedicine ; 129: 155661, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38677269

RESUMO

BACKGROUND: Gallbladder cancer (GBC) poses a significant risk to human health. Its development is influenced by numerous factors, particularly the homeostasis of reactive oxygen species (ROS) within cells. This homeostasis is crucial for tumor cell survival, and abnormal regulation of ROS is associated with the occurrence and progression of many cancers. Dihydrotanshinone I (DHT I), a biologically effective ingredient isolated from Salvia miltiorrhiza, has exhibited cytotoxic properties against various tumor cells by inducing apoptosis. However, the precise molecular mechanisms by which dht I exerts its cytotoxic effects remain unclear. PURPOSE: To explore the anti-tumor impact of dht I on GBC and elucidate the potential molecular mechanisms. METHODS: The proliferation of GBC cells, NOZ and SGC-996, was assessed using various assays, including CCK-8 assay, colony formation assay and EdU staining. We also examined cell apoptosis, cell cycle progression, ROS levels, and alterations in mitochondrial membrane potential to delve into the intricate molecular mechanism. Quantitative PCR (qPCR), immunofluorescence staining, and Western blotting were performed to evaluate target gene expression at both the mRNA and protein levels. The correlation between nuclear factor erythroid 2-related factor 2 (Nrf2) and kelch-like ECH-associated protein 1 (Keap1) were examined using co-immunoprecipitation. Finally, the in vivo effect of dht I was investigated using a xenograft model of gallbladder cancer in mice. RESULTS: Our research findings indicated that dht I exerted cytotoxic effects on GBC cells, including inhibiting proliferation, disrupting mitochondrial membrane potential, inducing oxidative stress and apoptosis. Our in vivo studies substantiated the inhibition of dht I on tumor growth in xenograft nude mice. Mechanistically, dht I primarily targeted Nrf2 by promoting Keap1 mediated Nrf2 degradation and inhibiting protein kinase C (PKC) induced Nrf2 phosphorylation. This leads to the suppression of Nrf2 nuclear translocation and reduction of its target gene expression. Moreover, Nrf2 overexpression effectively counteracted the anti-tumor effects of dht I, while Nrf2 knockdown significantly enhanced the inhibitory effect of dht I on GBC. Meanwhile, PKC inhibitors and nuclear import inhibitors increased the sensitivity of GBC cells to dht I treatment. Conversely, Nrf2 activators, proteasome inhibitors, antioxidants and PKC activators all antagonized dht I induced apoptosis and ROS generation in NOZ and SGC-996 cells. CONCLUSION: Our findings indicated that dht I inhibited the growth of GBC cells by regulating the Keap1-Nrf2 signaling pathway and Nrf2 phosphorylation. These insights provide a strong rationale for further investigation of dht I as a potential therapeutic agent for GBC treatment.


Assuntos
Apoptose , Proliferação de Células , Neoplasias da Vesícula Biliar , Proteína 1 Associada a ECH Semelhante a Kelch , Camundongos Nus , Fator 2 Relacionado a NF-E2 , Fenantrenos , Espécies Reativas de Oxigênio , Transdução de Sinais , Animais , Humanos , Camundongos , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Furanos/farmacologia , Neoplasias da Vesícula Biliar/tratamento farmacológico , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Fator 2 Relacionado a NF-E2/metabolismo , Fenantrenos/farmacologia , Fosforilação/efeitos dos fármacos , Quinonas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Salvia miltiorrhiza/química , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Heliyon ; 10(5): e27618, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38495177

RESUMO

Acyl-CoA thioesterase 4 (ACOT4) has been reported to be related to acetyl-CoA carboxylase activity regulation; However, its exact functions in liver lipid and glucose metabolism are still unclear. Here, we discovered explored the regulatory roles of ACOT4 in hepatic lipid and glucose metabolism in vitro. We found that the expression level of ACOT4 was significantly increased in the hepatic of db/db and ob/ob mice as well as obese mice fed a high fat diet. Adenovirus-mediated overexpression of ACOT4 promoted gluconeogenesis and high-glucose/high-insulin-induced lipid accumulation and impaired insulin sensitivity in primary mouse hepatocytes, whereas ACOT4 knockdown notably suppressed gluconeogenesis and decreased the triglycerides accumulation in hepatocytes. Furthermore, ACOT4 knockdown increased insulin-induced phosphorylation of AKT and GSK-3ß in primary mouse hepatocytes. Mechanistically, we found that upregulation of ACOT4 expression inhibited AMP-activated protein kinase (AMPK) activity, and its knockdown had the opposite effect. However, activator A769662 and inhibitor compound C of AMPK suppressed the impact of the change in ACOT4 expression on AMPK activity. Our data indicated that ACOT4 is related to hepatic glucose and lipid metabolism, primarily via the regulation of AMPK activity. In conclusion, ACOT4 is a potential target for the therapy of non-alcoholic fatty liver (NAFLD) and type 2 diabetes.

3.
Immunobiology ; 229(1): 152778, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38159526

RESUMO

Mesencephalic astrocyte-derived neurotrophic factor (MANF) is an endoplasmic reticulum (ER) stress-induced protein, and it has been reported that ER stress and unfolded protein response (UPR) are closely related to the immune system. The spleen is an important immune organ and we have shown in our previous research that MANF is expressed in human spleen tissues. However, there have been limited studies about the effect of MANF on spleen development. In this study, we detected MANF expression in spleen tissues and found that MANF was expressed in the red pulp and marginal zone. Additionally, MANF was localized in the CD68+ and CD138+ cells of adult rat spleen tissues, but not in the CD3+ cells. We performed immunohistochemical staining to detect MANF expression in the spleen tissues of rats that were different ages, and we found that MANF+ cells were localized together in the spleen tissues of rats that were 1-4 weeks old. MANF was also expressed in CD68+ cells in the spleen tissues of rats and mice. Furthermore, we found that MANF deficiency inhibited white pulp development in MANF knockout mice, thus indicating that MANF played an important role in the white pulp development of rodent spleen tissues.


Assuntos
Astrócitos , Baço , Animais , Humanos , Camundongos , Ratos , Astrócitos/metabolismo , Estresse do Retículo Endoplasmático , Camundongos Knockout , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Fatores de Crescimento Neural/farmacologia , Baço/metabolismo , Resposta a Proteínas não Dobradas
4.
bioRxiv ; 2023 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-37645772

RESUMO

In the brain, connectivity determines function. Neurons in the parabrachial nucleus (PB) relay diverse information to widespread brain regions, but the connections and functions of PB neurons that express Nps (neuropeptide S) remain mysterious. Here, we use Cre-dependent anterograde tracing and whole-brain analysis to map their output connections. While many other PB neurons project ascending axons through the central tegmental tract, NPS axons reach the forebrain via distinct periventricular and ventral pathways. Along the periventricular pathway, NPS axons target the tectal longitudinal column and periaqueductal gray then continue rostrally to target the paraventricular nucleus of the thalamus. Along the ventral pathway, NPS axons blanket much of the hypothalamus but avoid the ventromedial and mammillary nuclei. They also project prominently to the ventral bed nucleus of the stria terminalis, A13 cell group, and magnocellular subparafasciular nucleus. In the hindbrain, NPS axons have fewer descending projections, targeting primarily the superior salivatory nucleus, nucleus of the lateral lemniscus, and periolivary region. Combined with what is known about NPS and its receptor, the output pattern of Nps-expressing neurons in the PB region predicts a role in threat response and circadian behavior.

5.
Adv Sci (Weinh) ; 10(28): e2301166, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37566761

RESUMO

Most nonalcoholic steatohepatitis (NASH) patients develop severe fibrosis through extracellular matrix (ECM) accumulation, which can lead to hepatocellular carcinoma (HCC). Fibroblast growth factor 9 (FGF9) is involved in serial types of cancer; however, the specific role of FGF9 in NASH-driven HCC is not fully understood. This study finds that FGF9 is increased in patients with NASH-associated HCC. Furthermore, NASH-driven HCC mice models by feeding wildtype mice with high-fat/high-cholesterol (HFHC) diet and low dose carbon tetrachloride (CCl4 ) treatment is established; and identified that hepatic FGF9 is increased; with severe fibrosis. Additionally, AAV-mediated knockdown of FGF9 reduced the hepatic tumor burden of NASH-driven HCC mice models. Hepatocyte-specific FGF9 transgenic mice (FGF9Alb ) fed with a HFHC diet without CCl4 treatment exhibited an increased hepatic ECM and tumor burden. However, XAV-939 treatment blocked ECM accumulation and NASH-driven HCC in FGF9Alb mice fed with HFHC diet. Molecular mechanism studies show that FGF9 stimulated the expression of ECM related genes in a ß-catenin dependent manner; and FGF9 exerts its effect on ß-catenin stability via the ERK1/2-GSK-3ß signaling pathway. In summary, the data provides evidence for the critical role of FGF9 in NASH-driven HCC pathogenesis; wherein it promotes the tumors formation through the ECM pathway.

6.
Acta Pharmacol Sin ; 44(7): 1416-1428, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36721007

RESUMO

Non-alcoholic fatty liver disease (NAFLD) is a major health concern worldwide, and the incidence of metabolic disorders associated with NAFLD is rapidly increasing because of the obesity epidemic. There are currently no approved drugs that prevent or treat NAFLD. Recent evidence shows that bavachin, a flavonoid isolated from the seeds and fruits of Psoralea corylifolia L., increases the transcriptional activity of PPARγ and insulin sensitivity during preadipocyte differentiation, but the effect of bavachin on glucose and lipid metabolism remains unclear. In the current study we investigated the effects of bavachin on obesity-associated NAFLD in vivo and in vitro. In mouse primary hepatocytes and Huh7 cells, treatment with bavachin (20 µM) significantly suppressed PA/OA or high glucose/high insulin-induced increases in the expression of fatty acid synthesis-related genes and the number and size of lipid droplets. Furthermore, bavachin treatment markedly elevated the phosphorylation levels of AKT and GSK-3ß, improving the insulin signaling activity in the cells. In HFD-induced obese mice, administration of bavachin (30 mg/kg, i.p. every other day for 8 weeks) efficiently attenuated the increases in body weight, liver weight, blood glucose, and liver and serum triglyceride contents. Moreover, bavachin administration significantly alleviated hepatic inflammation and ameliorated HFD-induced glucose intolerance and insulin resistance. We demonstrated that bavachin protected against HFD-induced obesity by inducing fat thermogenesis and browning subcutaneous white adipose tissue (subWAT). We revealed that bavachin repressed the expression of lipid synthesis genes in the liver of obese mice, while promoting the expression of thermogenesis, browning, and mitochondrial respiration-related genes in subWAT and brown adipose tissue (BAT) in the mice. In conclusion, bavachin attenuates hepatic steatosis and obesity by repressing de novo lipogenesis, inducing fat thermogenesis and browning subWAT, suggesting that bavachin is a potential drug for NAFLD therapy.


Assuntos
Resistência à Insulina , Hepatopatia Gordurosa não Alcoólica , Animais , Camundongos , Hepatopatia Gordurosa não Alcoólica/metabolismo , Camundongos Obesos , Glicogênio Sintase Quinase 3 beta/metabolismo , Fígado/metabolismo , Obesidade/complicações , Obesidade/tratamento farmacológico , Obesidade/genética , Flavonoides/farmacologia , Dieta , Glucose/metabolismo , Insulina/metabolismo , Dieta Hiperlipídica , Camundongos Endogâmicos C57BL
7.
Am J Transl Res ; 14(10): 7552-7565, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36398234

RESUMO

BACKGROUND: Long term peritoneal dialysis leads to peritoneal epithelial-mesenchymal transformation (EMT), angiogenesis, and ultrafiltration failure. Although recent evidence suggests that inhibiting STAT3 (signal transducer and activator of transcription 3) can prevent kidney fibrosis, and that STAT3 can enhance glucose metabolism, the effect of STAT3 in peritoneal fibrosis (PF) has not been clarified. METHODS: Our study determined the effects of STAT3 on EMT and key glycolysis enzymes in mesothelial HMrSV5 cells by knockdown and overexpression of STAT3. In addition, we established a rat PF model to examine the role of pharmacologic inhibition of STAT3 or 6-phosphofructo-2-kinase/fructose-2, 6-bisphosphatase 3 (PFKFB3) in this process. RESULTS: High glucose (HG) caused the upregulation of α-smooth muscle actin and transforming growth factor beta 1 and the downregulation of E-cadherin, and induced STAT3 activation in HMrSV5 cells. In addition, HMrSV5 cells cultured in high glucose showed high expression of key glycolysis enzymes, which could be inhibited by STAT3 siRNA. Furthermore, treating mesothelial cells with 3PO, the PFKFB3 inhibitor, could attenuate high glucose-induced EMT. Moreover, daily administration of dialysis fluid could induce peritoneal fibrosis. The peritoneal fibrosis was accompanied by enhanced phosphorylation of STAT3 and the upregulation of PFKFB3. The administration of BP-1-102 or 3PO prevented fibrosis and inhibited angiogenesis in PF rats. CONCLUSIONS: si-STAT3 attenuated the HG-induced EMT and hyperglycolysis, and the overexpression of STAT3 could induce EMT in HMrSV5 cells. 3PO could markedly attenuate HG-induced EMT by decreasing PFKEB3 in HMrSV5 cells. In addition, we demonstrated that inhibiting STAT3 signaling or peritoneal hyperglycolysis could attenuate peritoneal fibrosis and angiogenesis in vivo. Our findings linked the STAT3/PFKFB3 signaling to the development of PF. HG/STAT3/PFKFB3 might promote the progression of PF through regulating profibrosis and angiogenesis.

8.
J Comp Neurol ; 530(18): 3157-3178, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36036349

RESUMO

Neuropeptide S (NPS) increases wakefulness. A small number of neurons in the brainstem express Nps. These neurons are located in or near the parabrachial nucleus (PB), but we know very little about their ontogeny, connectivity, and function. To identify Nps-expressing neurons within the molecular framework of the PB region, we used in situ hybridization, immunofluorescence, and Cre-reporter labeling in mice. The primary concentration of Nps-expressing neurons borders the lateral lemniscus at far-rostral levels of the lateral PB. Caudal to this main cluster, Nps-expressing neurons scatter through the PB and form a secondary concentration medial to the locus coeruleus (LC). Most Nps-expressing neurons in the PB region are Atoh1-derived, Foxp2-expressing, and mutually exclusive with neurons expressing Calca or Lmx1b. Among Foxp2-expressing PB neurons, those expressing Nps are distinct from intermingled subsets expressing Cck or Pdyn. Examining Nps Cre-reporter expression throughout the brain identified novel populations of neurons in the nucleus incertus, anterior hypothalamus, and lateral habenula. This information will help focus experimental questions about the connectivity and function of NPS neurons.


Assuntos
Neurônios , Núcleos Parabraquiais , Animais , Camundongos , Neurônios/metabolismo , Encéfalo/metabolismo , Hibridização In Situ , Tronco Encefálico
9.
J Comp Neurol ; 530(10): 1658-1699, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35134251

RESUMO

Diverse neurons in the parabrachial nucleus (PB) communicate with widespread brain regions. Despite evidence linking them to a variety of homeostatic functions, it remains difficult to determine which PB neurons influence which functions because their subpopulations intermingle extensively. An improved framework for identifying these intermingled subpopulations would help advance our understanding of neural circuit functions linked to this region. Here, we present the foundation of a developmental-genetic ontology that classifies PB neurons based on their intrinsic, molecular features. By combining transcription factor labeling with Cre fate-mapping, we find that the PB is a blend of two, developmentally distinct macropopulations of glutamatergic neurons. Neurons in the first macropopulation express Lmx1b (and, to a lesser extent, Lmx1a) and are mutually exclusive with those in a second macropopulation, which derive from precursors expressing Atoh1. This second, Atoh1-derived macropopulation includes many Foxp2-expressing neurons, but Foxp2 also identifies a subset of Lmx1b-expressing neurons in the Kölliker-Fuse nucleus (KF) and a population of GABAergic neurons ventrolateral to the PB ("caudal KF"). Immediately ventral to the PB, Phox2b-expressing glutamatergic neurons (some coexpressing Lmx1b) occupy the KF, supratrigeminal nucleus, and reticular formation. We show that this molecular framework organizes subsidiary patterns of adult gene expression (including Satb2, Calca, Grp, and Pdyn) and predicts output projections to the amygdala (Lmx1b), hypothalamus (Atoh1), and hindbrain (Phox2b/Lmx1b). Using this molecular ontology to organize, interpret, and communicate PB-related information could accelerate the translation of experimental findings from animal models to human patients.


Assuntos
Núcleo de Kölliker-Fuse , Núcleos Parabraquiais , Animais , Encéfalo/metabolismo , Neurônios GABAérgicos/metabolismo , Humanos , Hipotálamo/metabolismo , Ponte/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
11.
Mol Med Rep ; 24(6)2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34590153

RESUMO

The morbidity and mortality of pancreatic cancer have been continuously increasing, causing seven deaths per 100,000 individuals/year. At present, effective therapies are severely lacking, thus, highlighting the importance of developing novel therapeutic approaches. The present study aimed to investigate the inhibitory roles of the 2,3­oxidosqualene cyclase inhibitor, RO 48­8071 (RO), on pancreatic ductal adenocarcinoma. RO was used to treat the pancreatic cancer cell line (PANC­1) in vitro to examine the effects of RO on cell viability, as well as to determine its potential molecular mechanism. Moreover, experiments in a xenograft model of subcutaneous tumors generated by injecting PANC­1 cells hypodermically into nude mice were performed to observe the inhibition of RO on tumor growth. It was found that RO inhibited PANC­1 cell viability when treatment was given for 24, 48 and 72 h. The in vivo study demonstrated that RO markedly inhibited subcutaneous tumor growth in nude mice. Further studies revealed that RO could induce cell cycle arrest in the G1 phase by regulating p27, cyclin B1 and cyclin E expression to inhibit PANC­1 cell viability. Moreover, RO inactivated the JNK and ERK MAPK signaling pathway by decreasing the phosphorylation levels of JNK and ERK. Collectively, the present study demonstrated that RO served anti­pancreatic cancer roles in vitro and in vivo, which may provide new ideas and facilitate the development of novel treatment options for pancreatic cancer.


Assuntos
Benzofenonas/farmacologia , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Metabolismo dos Lipídeos/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Colesterol/biossíntese , Ciclina B1/metabolismo , Ciclina E/metabolismo , Humanos , Masculino , Camundongos , Camundongos Nus , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Fosforilação , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Int J Oncol ; 59(2)2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34195850

RESUMO

Pancreatic cancer ranks seventh in terms of cancer­related mortality in men and women worldwide, where the most common subtype is pancreatic ductal adenocarcinoma (PDAC). To date, the pathogenesis of PDAC remains incompletely understood and the prognosis of PDAC is poor. In the present study, the expression of interleukin­28 receptor α subunit (IL­28RA) in PDAC tissues was detected using immunofluorescence staining and western blotting. IL­28RA recombinant plasmids and control pCMV6­entrymammalian expression plasmid, short hairpin (sh)IL­28RA plasmids and control pRS scrambled shRNA vector purchased were used to produce stably transfected PANC­1 cells overexpressing IL­28RA or with IL­28RA expression knocked down. MTS assays were used to measure cell viability and wound healing assay was used to assess the cell migratory ability in vitro. Flow cytometry analysis was performed to determine the proportion of cells in each phase of the cell cycle whereas total protein and phosphorylated protein levels were assessed using western blotting. Xenograft models of subcutaneous tumors were established by injecting PANC­1 cells hypodermically into nude mice to investigate the effect of IL­28RA on tumorigenesis and tumor growth. The results showed that the expression of IL­28RA in PDAC tissues was lower compared with that in normal tissues. IL­28RA overexpression in vitro resulted in the activation of the IL­28RA pathway, which reduced cell viability and decreased the proportion of cells in the G2/M phase by reducing cyclin B1 expression. In addition, IL­28RA overexpression inhibited migration of PDAC cells. By contrast, an increased proportion of cells in G2/M phase, upregulated cyclin B1 expression and enhanced cell viability and migratory ability along with inhibition of the IL­28RA pathway were observed in PANC­1 cells following IL­28RA knockdown. The inhibitory effect of IL­28RA was observed by tumor size in a nude mouse model induced by PANC­1 cells with stable IL­28RA overexpression or knockdown. The tumor size induced by PANC­1 cells with stable IL­28RA overexpression were smaller, whilst larger tumors induced by PANC­1 cells were observed following stable IL­28RA knockdown, when compared to control. Further studies showed that the effect of IL­28RA on PDAC cells was exerted by regulating the phosphorylation levels of STAT1 and AKT. In conclusion, lower IL­28RA expression may contribute to the pathogenesis of PDAC, where results from the present may provide further insights into the progression of PDAC, in addition to highlighting potentially novel therapeutic targets for this disease.


Assuntos
Carcinoma Ductal Pancreático/patologia , Regulação para Baixo , Neoplasias Pancreáticas/patologia , Receptores de Interferon/genética , Receptores de Interferon/metabolismo , Idoso , Animais , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Ciclo Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Transplante de Neoplasias , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Prognóstico
13.
Can J Physiol Pharmacol ; 99(10): 1079-1087, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33915055

RESUMO

Chronic cerebral hypoperfusion (CCH), as a critical factor of chronic cerebrovascular diseases, has greatly influenced the health of patients with vascular dementia. Vitexin, a flavone C-glycoside (apigenin-8-C-ß-D-glucopyranoside) that belongs to the flavone subclass of flavonoids, has been shown to possess antioxidant and anti-ischemic properties; however, the putative protective effects of vitexin on the CCH need further investigation. In the current study, the role of vitexin and its underlying mechanism were investigated with permanent bilateral common carotid artery occlusion (2VO) in rats as well as mouse hippocampal neuronal (HT22) cells with oxygen and glucose deprivation/reoxygenation (OGD/R) injury model. The results demonstrated that vitexin improved cognitive dysfunction as well as alleviated pathological neuronal damage in hematoxylin plus eosin (HE) and TUNEL results. The decreased levels of exchange protein directly activated by cAMP 1 (Epac1), Epac2, Ras-associated protein 1 (Rap1), and phospho-extracellular signal-regulated kinase (p-ERK) were reversed by vitexin in rats with CCH. Furthermore, this study indicated that vitexin alleviated CCH-induced inflammation injuries by reducing the expression of NOD-like receptor 3 (NLRP3), caspase-1, interleukin 1ß (IL-1ß), IL-6, and cleaved caspase-3. In vitro, vitexin increased the expression of Epac1 and Epac2, decreased the activation of the NLRP3-mediated inflammation, and improved cell viability. Taken together, our findings suggest that vitexin can reduce the degree of the progressing pathological damage in the cortex and hippocampus and inhibit further deterioration of cognitive function in rats with CCH. Epac and NLRP3 can be regulated by vitexin in vivo and in vitro, which provides enlightenment for the protection of CCH injury.


Assuntos
Apigenina/farmacologia , Transtornos Cerebrovasculares/tratamento farmacológico , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Hipocampo/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Neurônios/efeitos dos fármacos , Animais , Circulação Cerebrovascular , Transtornos Cerebrovasculares/metabolismo , Transtornos Cerebrovasculares/patologia , Doença Crônica , Transtornos Cognitivos/metabolismo , Transtornos Cognitivos/patologia , Transtornos Cognitivos/prevenção & controle , Fatores de Troca do Nucleotídeo Guanina/genética , Hipocampo/metabolismo , Hipocampo/patologia , Masculino , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Neurônios/metabolismo , Neurônios/patologia , Ratos , Ratos Sprague-Dawley
14.
J Comp Neurol ; 529(11): 2911-2957, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-33715169

RESUMO

The parabrachial nucleus (PB) is composed of glutamatergic neurons at the midbrain-hindbrain junction. These neurons form many subpopulations, one of which expresses Calca, which encodes the neuropeptide calcitonin gene-related peptide (CGRP). This Calca-expressing subpopulation has been implicated in a variety of homeostatic functions, but the overall distribution of Calca-expressing neurons in this region remains unclear. Also, while previous studies in rats and mice have identified output projections from CGRP-immunoreactive or Calca-expressing neurons, we lack a comprehensive understanding of their efferent projections. We began by identifying neurons with Calca mRNA and CGRP immunoreactivity in and around the PB, including populations in the locus coeruleus and motor trigeminal nucleus. Calca-expressing neurons in the PB prominently express the mu opioid receptor (Oprm1) and are distinct from neighboring neurons that express Foxp2 and Pdyn. Next, we used Cre-dependent anterograde tracing with synaptophysin-mCherry to map the efferent projections of these neurons. Calca-expressing PB neurons heavily target subregions of the amygdala, bed nucleus of the stria terminalis, basal forebrain, thalamic intralaminar and ventral posterior parvicellular nuclei, and hindbrain, in different patterns depending on the injection site location within the PB region. Retrograde axonal tracing revealed that the previously unreported hindbrain projections arise from a rostral-ventral subset of CGRP/Calca neurons. Finally, we show that these efferent projections of Calca-expressing neurons are distinct from those of neighboring PB neurons that express Pdyn. This information provides a detailed neuroanatomical framework for interpreting experimental work involving CGRP/Calca-expressing neurons and opioid action in the PB region.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina/biossíntese , Neurônios Eferentes/metabolismo , Núcleos Parabraquiais/metabolismo , Animais , Peptídeo Relacionado com Gene de Calcitonina/genética , Vias Eferentes/química , Vias Eferentes/metabolismo , Feminino , Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/química , Neurônios/metabolismo , Neurônios Eferentes/química , Núcleos Parabraquiais/química
15.
Int J Clin Exp Pathol ; 14(2): 221-229, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33564354

RESUMO

OBJECTIVE: This study aimed to explore the relationship between diabetic xerostomia and changes in aquaporin-1 (AQP1), aquaporin-5 (AQP5), and aquaporin-8 (AQP8) expression in the submandibular glands (SMGs), to further study the pathogenesis of diabetic xerostomia and to observe the therapeutic effect of insulin (INS). METHODS: Thirty SD rats were randomized equally into 3 groups: control group, diabetic model (DM) group and insulin (INS) group (n=10, respectively). The control group received no treatment. DM group and INS group were induced by a high-fat diet and streptozotocin intraperitoneal injection. After establishment of a diabetic rat model, the rats in INS group were treated with insulin. Then all rats were fed continuously with ordinary diet for 2 months. H&E staining was used to describe morphologic changes in the SMGs of rats. Immunohistochemistry was used to analyze the expressions and localization of AQP1, AQP5, and AQP8 in the SMGs. Computer image analysis was used to detect the mean optical density (MOD) values of AQP1, AQP5, and AQP8 expression, and changes in the diameters of acini and ducts. RESULTS: The acini were mildly atrophied and the acinar cells were rearranged in an irregular way. The morphology of insulin-administered diabetic SMGs was similar to that of the control group. The acinar average circumference and GCT average diameter in DM group were significantly reduced (P<0.05). The acinar average circumference and GCT average diameter of INS group were significantly increased (P<0.05). The expressions of AQP1, AQP5, and AQP8 were significantly reduced in DM group (P<0.05). The expressions of AQP1, AQP5, and AQP8 in INS group were significantly increased (P<0.05). CONCLUSION: The decreased expressions of AQP1, AQP5, and AQP8 led to decreased salivary secretion of SMGs in diabetic rats, which may be involved in the pathogenesis of diabetic xerostomia. Insulin could up-regulate the expressions of AQP1, AQP5 and AQP8, and play a protective role in the secretory function of diabetic SMGs.

16.
J Comp Neurol ; 529(4): 657-693, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32621762

RESUMO

The parabrachial nucleus (PB) is a complex structure located at the junction of the midbrain and hindbrain. Its neurons have diverse genetic profiles and influence a variety of homeostatic functions. While its cytoarchitecture and overall efferent projections are known, we lack comprehensive information on the projection patterns of specific neuronal subtypes in the PB. In this study, we compared the projection patterns of glutamatergic neurons here with a subpopulation expressing the transcription factor Foxp2 and a further subpopulation expressing the neuropeptide Pdyn. To do this, we injected an AAV into the PB region to deliver a Cre-dependent anterograde tracer (synaptophysin-mCherry) in three different strains of Cre-driver mice. We then analyzed 147 neuroanatomical regions for labeled boutons in every brain (n = 11). Overall, glutamatergic neurons in the PB region project to a wide variety of sites in the cerebral cortex, basal forebrain, bed nucleus of the stria terminalis, amygdala, diencephalon, and brainstem. Foxp2 and Pdyn subpopulations project heavily to the hypothalamus, but not to the cortex, basal forebrain, or amygdala. Among the few differences between Foxp2 and Pdyn cases was a notable lack of Pdyn projections to the ventromedial hypothalamic nucleus. Our results indicate that genetic identity determines connectivity (and therefore, function), providing a framework for mapping all PB output projections based on the genetic identity of its neurons. Using genetic markers to systematically classify PB neurons and their efferent projections will enhance the translation of research findings from experimental animals to humans.


Assuntos
Encefalinas/biossíntese , Fatores de Transcrição Forkhead/biossíntese , Núcleos Parabraquiais/metabolismo , Precursores de Proteínas/biossíntese , Proteínas Repressoras/biossíntese , Proteína Vesicular 2 de Transporte de Glutamato/biossíntese , Animais , Tronco Encefálico/química , Tronco Encefálico/metabolismo , Córtex Cerebral/química , Córtex Cerebral/metabolismo , Vias Eferentes/química , Vias Eferentes/metabolismo , Encefalinas/análise , Encefalinas/genética , Feminino , Fatores de Transcrição Forkhead/análise , Fatores de Transcrição Forkhead/genética , Hipotálamo/química , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Núcleos Parabraquiais/química , Precursores de Proteínas/análise , Precursores de Proteínas/genética , Proteínas Repressoras/análise , Proteínas Repressoras/genética , Tálamo/química , Tálamo/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/análise , Proteína Vesicular 2 de Transporte de Glutamato/genética
17.
Cell Biol Int ; 44(7): 1481-1490, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32181963

RESUMO

Pyroptosis is a form of cell death that is uniquely dependent on caspase-1. Pyroptosis involved in oxidized low-density lipoprotein (ox-LDL)-induced human macrophage death through the promotion of caspase-1 activation is important for the formation of unstable plaques in atherosclerosis. The mitochondrial outer membrane protein NIX directly interacts with microtubule-associated protein 1 light chain 3 (LC3). Although we previously showed that NIX-mediated mitochondrial autophagy is involved in the clearance of damaged mitochondria, how NIX contributes to ox-LDL-induced macrophage pyroptosis remains unknown. Here, immunoperoxidase staining Nix expression decreased in human atherosclerosis. When we silenced NIX expression in murine macrophage cell, active caspase-1, and mature interleukin-1ß expression levels were increased and LC3 was reduced. In addition, LDH release and acridine orange and ethidium bromide staining indicated that damage to macrophage cell membranes induced by ox-LDL was substantially worse. Moreover, intracellular reactive oxygen species and NLRP3 inflammasome levels increased. Taken together, these results demonstrated that NIX inhibits ox-LDL-induced macrophage pyroptosis via autophagy in atherosclerosis.


Assuntos
Aterosclerose/patologia , Autofagia/efeitos dos fármacos , Lipoproteínas LDL/farmacologia , Macrófagos/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Aterosclerose/induzido quimicamente , Aterosclerose/metabolismo , Morte Celular/efeitos dos fármacos , Humanos , Inflamassomos/efeitos dos fármacos , Inflamassomos/metabolismo , Lipoproteínas LDL/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Mitocôndrias/metabolismo
18.
Exp Mol Pathol ; 114: 104402, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32061942

RESUMO

The aim of this study was to investigate the roles of CD4+ T cells and transforming growth factor beta (TGFß1) in the pathological process of valvular hyperblastosis and fibrosis of patients with rheumatic heart disease (RHD). A total of 151 patients were enrolled, among whom, 78 patients were with RHD, and 73 were age and gender matched RHD negative patients. Blood samples and valve specimens were collected for analysis. Pathological changes and collagen fibers contents of valves were analyzed using HE and Masson staining. Percentage of peripheral blood CD4+ T cells was tested through flow cytometry. TGFß1 level in serum were identified by ELISA. CD4+ T cells infiltration and expression of TGFß1, p-p38, p-JNK, p-ERK in valves were detected by immunohistochemistry. The mRNA and protein levels of p38, JNK, ERK, TGFß1, I-collagen and α-SMA were detected by qRT-PCR and western blotting, respectively. The heart valve tissues of RHD patients showed higher degrees of fibrosis, calcification and lymphocytes infiltration, which were mainly CD4+ T cells. In addition, compared with control group, RHD patients had more total CD4+ T cells in peripheral blood and valve tissues. Expression of TGFß1, phosphorylation of JNK and p38, and synthesis of I-collagen in valve tissues of RHD patients were also significantly increased. Furthermore, we found a strong positive correlation between TGFß1 expression and phosphorylation of JNK and p38. CD4+ T cells, and fibrogenic cytokine TGFß1, which activate the intracellular MAPK signaling pathway may participate in the fibrosis of heart valve in RHD patients.


Assuntos
Doenças das Valvas Cardíacas/genética , Estenose da Valva Mitral/genética , Cardiopatia Reumática/genética , Fator de Crescimento Transformador beta1/genética , Adulto , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/sangue , MAP Quinases Reguladas por Sinal Extracelular/genética , Feminino , Fibrose/sangue , Fibrose/genética , Fibrose/patologia , Regulação da Expressão Gênica/genética , Doenças das Valvas Cardíacas/sangue , Doenças das Valvas Cardíacas/patologia , Humanos , MAP Quinase Quinase 4/sangue , MAP Quinase Quinase 4/genética , Sistema de Sinalização das MAP Quinases/genética , Masculino , Pessoa de Meia-Idade , Estenose da Valva Mitral/sangue , Estenose da Valva Mitral/patologia , Cardiopatia Reumática/sangue , Cardiopatia Reumática/patologia , Fator de Crescimento Transformador beta1/sangue , Proteínas Quinases p38 Ativadas por Mitógeno/sangue , Proteínas Quinases p38 Ativadas por Mitógeno/genética
19.
Biomed Pharmacother ; 124: 109849, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31972356

RESUMO

Vitexin (VT) is a main bioactive flavonoid compound derived from the dried leaf of hawthorn (Crataegus pinnatifida), a widely used Chinese traditional folk medicine. Recent studies have shown that vitexin presents cardioprotective effects in vivo and in vitro. Mitochondrial dysfunction is a salient feature of myocardial ischemia/reperfusion (I/R) injury (MIRI), but the potential mechanism is still unclear. This study investigated the cardioprotective effect of vitexin against MIRI and its possible mechanism. Isolated SD rat hearts were subjected to MIRI in a Langendorff perfusion system, and H9c2 cells were subjected to hypoxia/reoxygenation (H/R) in vitro. Ex vivo experiments showed improved left ventricular function and reduced infarct size in the vitexin group. Transmission electron microscopy showed that I/R caused outer mitochondrial membrane rupture, cristae disappearance and vacuolation, while vitexin reduced mitochondrial damage and ultimately reduced cardiomyocyte apoptosis. In vitro, vitexin protected H9c2 cells from H/R-induced mitochondrial dysfunction, significantly reducing ROS levels; improving mitochondrial activity, mitochondrial membrane potential and ATP content; markedly increasing MFN2 expression and reducing the recruitment of Drp1 in mitochondria. These results suggest a new protective mechanism of vitexin for ischemic heart disease treatment.


Assuntos
Apigenina/farmacologia , Mitocôndrias/efeitos dos fármacos , Dinâmica Mitocondrial/efeitos dos fármacos , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/patologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo
20.
Aging Cell ; 19(2): e13099, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31903715

RESUMO

Cancer is an age-associated disease, potentially related to the altered immune system of elderly individuals. However, cancer has gradually decreased incidence in the eldest globally such as the most common lung cancer, the mechanisms of which remain to be elucidated. In this study, it was found that the number of lung-resident γδT cells was significantly increased with altered gene expression in aged mice (20-24 months) versus young mice (10-16 weeks). Aged lung Vγ4+ and Vγ6+ γδT cells predominantly produced interleukin-17A (IL-17A), resulting in increased levels in the serum and lungs. Moreover, the aged mice exhibited smaller tumors and reduced numbers of tumor foci in the lungs after challenge with intravenous injection of B16/F10 melanoma cells compared with the young mice. Aged lung Vγ4+ and Vγ6+ γδT cells were highly cytotoxic to B16/F10 melanoma cells with higher expression levels of CD103. The markedly longer survival of the challenged aged mice was dependent on γδT17 cells, since neutralization of IL-17A or depletion of indicated γδT cells significantly shortened the survival time. Consistently, supplementation of IL-17A significantly enhanced the survival time of young mice with lung melanoma. Furthermore, the anti-tumor activity of aged lung γδT17 cells was not affected by alterations in the load and composition of commensal microbiota, as demonstrated through co-housing of the aged and young mice. Intrinsically altered lung γδT17 cells underlying age-dependent changes control lung melanoma, which will help to better understand the lung cancer progression in the elderly and the potential use of γδT17 cells in anti-tumor immunotherapy.


Assuntos
Envelhecimento/imunologia , Interleucina-17/metabolismo , Linfócitos Intraepiteliais/imunologia , Neoplasias Pulmonares/imunologia , Pulmão/imunologia , Melanoma Experimental/imunologia , Idoso , Envelhecimento/patologia , Animais , Antígenos CD/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/imunologia , Ontologia Genética , Humanos , Imunoterapia , Cadeias alfa de Integrinas/metabolismo , Interleucina-17/farmacologia , Interleucina-17/uso terapêutico , Pulmão/citologia , Pulmão/patologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Microbiota/imunologia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...