Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Mycotoxin Res ; 40(2): 255-268, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38400893

RESUMO

Aflatoxin B1 (AFB1) is a widespread toxic contamination in feed for animals. The primary active component of turmeric, curcumin (Cur), is an antioxidant and an anti-inflammatory. However, it is yet unknown how AFB1 affects the intestinal epithelial barrier and whether Cur acts as a protective mechanism when exposed to AFB1. Here, we explored the mechanism of AFB1-induced intestinal injury from intestinal epithelial barrier, inflammation, pyroptosis, and intestinal flora, and evaluated the protective role of Cur. We found that AFB1 caused weight loss and intestinal morphological damage that is mainly characterized by shortened intestinal villi, deepened crypts, and damaged intestinal epithelium. Exposure to AFB1 decreased the expression of Claudin-1, MUC2, ZO-1, and Occludin and increased the expression of pyroptosis-related factors (NLRP3, GSDMD, Caspase-1, IL-1ß, and IL-18) and inflammation-related factors (TLR4, NF-κB, IκB, IFN-γ, and TNF-α). Furthermore, ileal gut microbiota was altered, and simultaneously, the Lactobacillus abundance was decreased. The gut microbiota interacts with a wide range of physiologic functions and disease development in the host through its metabolites, and disturbances in gut microbial metabolism can cause functional impairment of the ileum. Meanwhile, Cur can ameliorate histological ileum injuries and intestinal flora disturbance caused by AFB1. We found that Cur reversed the effects of AFB1 through modulating both NLRP3 inflammasome and the TLR4/NF-κB signaling pathway. In conclusion, AFB1 can induce inflammatory damage and pyroptosis in duck ileum, while Cur has obviously protective effects on all the above damages.


Assuntos
Aflatoxina B1 , Curcumina , Patos , Íleo , Inflamassomos , NF-kappa B , Proteína 3 que Contém Domínio de Pirina da Família NLR , Transdução de Sinais , Receptor 4 Toll-Like , Animais , Aflatoxina B1/toxicidade , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/metabolismo , Curcumina/farmacologia , Inflamassomos/metabolismo , Íleo/efeitos dos fármacos , Íleo/patologia , Microbioma Gastrointestinal/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Mucosa Intestinal/microbiologia
2.
Artigo em Inglês | MEDLINE | ID: mdl-38061615

RESUMO

Aflatoxin B1 (AFB1) is the most prevalent and toxic class of aflatoxins, which is considered a significant risk factor for food safety. Curcumin, a phytoconstituent with anti-inflammatory and antioxidant properties, has potential therapeutic value for intestinal inflammatory diseases. In this study, the duckling model susceptible to AFB1 was selected for toxicity testing, aiming to explore the effect of curcumin on AFB1 enterotoxicity and its possible mechanism of action. The results showed that curcumin promoted the growth and development of ducklings and mitigated the changes in morphology and permeability serological index (DAO and D-LA) after AFB1 exposure. Curcumin also mitigated AFB1-induced oxidative stress by activating the Nrf2 pathway, and ameliorated intestinal inflammation by inhibiting the NF-κB/IκB signaling pathway and boosting intestinal autophagy. In terms of gut flora and their metabolites, we found that curcumin supplementation significantly increased the intestinal flora's abundance index and diversity index compared to the AFB1 group, mitigating the decline in the abundance of Actinobacteria and the rise in that of harmful bacteria Clostridia. Furthermore, untargeted metabolomic analysis revealed that the protective effect of curcumin on the intestine was mainly through the regulation of AFB1-induced disorders of lipid metabolism, involving linoleic acid metabolism, α-linolenic acid metabolism, and glycerolipid metabolism. Overall, the enteroprotective effects of curcumin may be of significant value in the future for treating chronic AFB1 poisoning and also provide new therapeutic ideas for other mycotoxicosis.


Assuntos
Aflatoxina B1 , Curcumina , Animais , Aflatoxina B1/toxicidade , Curcumina/farmacologia , Patos/metabolismo , Multiômica , Fígado/metabolismo , Estresse Oxidativo , Intestinos
3.
J Trace Elem Med Biol ; 81: 127336, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37976960

RESUMO

BACKGROUND: Arsenic is a widely distributed ecotoxic pollutant that has been found to cause neurotoxicity in a variety of species. Gut-brain axis is a two-way information network between the gut microbiome and the brain, which is closely related to organismal health. However, the role of the gut-brain axis in arsenic-induced neurotoxicity remains largely unknown. METHODS: In order to explore whether there is a relationship between brain and gut microbiota of meat ducks, we performed molecular biological detection including RT-qPCR and Western blot, as well as morphological detection including, HE staining and immunohistochemistry. Meanwhile, intestinal contents were analyzed using 16 S ribosomal RNA gene sequencing and analysis RESULTS: In this study, we investigated whether arsenic trioxide (ATO) can activate the gut microbiome-brain axis to induce intestinal and brain injury. The results showed that ATO-exposure disrupted the diversity balance of intestinal microbiota and integrity and injured the intestinal structure. ATO-exposure also reduced the number of glycogen and goblet cells in the duodenum. In addition, exposure to ATO caused intestinal inflammatory injury by activating NF-κB signaling pathway and promoting the expression of its target genes. Meanwhile, the tight junction-related proteins (ZO-1, occludin) of gut and brain were reduced by ATO exposure. Furthermore, results also revealed that ATO-exposure induced brain injury, including neuronal cell vacuolization and reduced numbers of neuronal cells in the cortex and hippocampus. Remarkably, ATO-exposure also disrupted neurotransmitter levels. Additionally, our further molecular mechanism study revealed that ATO-exposure increased the expression of autophagy and apoptosis related mRNA and proteins levels in the brain tissues. CONCLUSION: Altogether, these findings provide a new insight into that ATO-exposure induced intestinal injury and aggravated neurotoxicity via the gut-brain axis.


Assuntos
Arsênio , Lesões Encefálicas , Animais , Arsênio/toxicidade , Patos , Eixo Encéfalo-Intestino , Trióxido de Arsênio/farmacologia , Encéfalo
4.
Biol Trace Elem Res ; 2023 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-38102534

RESUMO

Arsenic has been shown to be highly toxic and can cause liver damage. Previous studies have shown that arsenic causes severe liver damage and induces accumulation of reactive oxygen species (ROS). This study aimed to investigate the effects of ferroptosis on the liver in arsenic trioxide (ATO) and to explore the underlying mechanisms. We confirmed the hepatotoxic effects of arsenic by in vivo and in vitro experiments. After 28 days of administration of arsenic trioxide (4-mg/kg, 8-mg/kg) by gavage, chickens exhibited body weight loss and liver damage in a dose-dependent manner. In addition, in vivo and in vitro western blot and real-time fluorescence quantitative PCR analyses simultaneously indicated that ferroptosis might be the main pathway of arsenic-induced liver injury. Finally, Mito-TEMPO effectively eliminated the ROS accumulation in mitochondria, significantly attenuating the process of cellular ferroptosis. In summary, the hepatotoxic effects of arsenic are related to ferroptosis, and the hepatic ferroptosis process of arsenic is regulated by mitochondrial ROS (MtROS). Our study reveals new mechanisms of arsenic toxicity to the liver, which may deepen our understanding of arsenic toxicology.

5.
Mycotoxin Res ; 39(4): 437-451, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37782431

RESUMO

Aflatoxin B1 (AFB1), an extremely toxic mycotoxin that extensively contaminates feed and food worldwide, poses a major hazard to poultry and human health. Curcumin, a polyphenol derived from turmeric, has attracted great attention due to its wonderful antioxidant properties. Nevertheless, effects of curcumin on the kidneys of ducks exposed to AFB1 remain unclear. Additionally, the underlying mechanism between AFB1 and ferroptosis (based on excessive lipid peroxidation) has not been sufficiently elucidated. This study aimed to investigate the protective effects and potential mechanisms of curcumin against AFB1-induced nephrotoxicity in ducklings. The results indicated that curcumin alleviated AFB1-induced growth retardation and renal distorted structure in ducklings. Concurrently, curcumin inhibited AFB1-induced mitochondrial-mediated oxidative stress by reducing the expression levels of oxidative damage markers malondialdehyde (MDA) and 8-hydroxy-2 deoxyguanosine (8-OHdG) and improved the expression of mitochondria-related antioxidant enzymes and the Nrf2 pathway. Notably, curcumin attenuated iron accumulation in the kidney, inhibited ferritinophagy via the NCOA4 pathway, and balanced iron homeostasis, thereby alleviating AFB1-induced ferroptosis in the kidney. Collectively, our results suggest that curcumin alleviates AFB1-induced nephrotoxicity in ducks by inhibiting mitochondrial-mediated oxidative stress, ferritinophagy, and ferroptosis and provide new evidence for the mechanism of AFB1-induced nephrotoxicity in ducklings treated with curcumin.


Assuntos
Curcumina , Ferroptose , Animais , Humanos , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Aflatoxina B1/toxicidade , Aflatoxina B1/metabolismo , Patos/metabolismo , Curcumina/farmacologia , Estresse Oxidativo , Ferro/farmacologia
7.
Food Chem Toxicol ; 172: 113593, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36596445

RESUMO

Aflatoxin B1 (AFB1), as the most toxic secondary metabolite produced by Aspergillus flavus, is a serious threat to human and animal health. Curcumin, a polyphenol from the plant turmeric, has demonstrated unique anti-damage properties in several studies. But, its ability to alleviate AFB1-induced liver damage in ducks and the underlying mechanisms are not completely elucidated. In this study, we investigated the intervention of curcumin on AFB1-induced hepatotoxicity in ducks. Research data showed that the combination of curcumin and AFB1 alleviated oxidative stress, reduced malondialdehyde (MDA) accumulation and relieved hepatotoxicity after 28 days of treatment, compared with AFB1. Also, curcumin upregulated the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream antioxidant enzymes (SOD, HO-1), which enhanced the antioxidant capacity of the liver. In addition, curcumin inhibited AFB1-induced lysosomal damage in the liver, with the character of reduced lysosomal membrane permeabilization, restored autophagic flux, and promoted lysosomal biogenesis, thereby enhancing the self-protective capacity of the liver. In conclusion, our results suggest that curcumin alleviates AFB1-induced duck hepatotoxicity by inhibiting oxidative stress and lysosomal damage.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas , Curcumina , Animais , Aflatoxina B1/toxicidade , Aflatoxina B1/metabolismo , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Curcumina/farmacologia , Curcumina/metabolismo , Patos/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Estresse Oxidativo
8.
Chemosphere ; 300: 134566, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35413363

RESUMO

The ecological risks caused by the coexistence of pollutants such as arsenic (As) and polystyrene-nanoplastics (PSNPs) in the environment have become a non-negligible problem. However, the effects of As and PSNPs co-exposure on mammals and the underlying toxicity mechanisms have remained unclear. Therefore, the present study established mouse models of As and/or PSNPS exposure to systematically analyze the underlying role of autophagy, apoptosis and pyroptosis in hepatotoxicity induced by co-exposure of As and PSNPs. Our findings demonstrated for the first time that mice co-exposure to As and PSNPs displayed significant pathological changes in the liver, while exposure to As or PSNPs alone did not produce significant toxic effects. More importantly, As and PSNPs co-exposure activated excessive autophagy through altered expression levels of PI3K, mTOR, Beclin-1, ATG5, LC3 and P62. Meanwhile, co-treatment with As and PSNPs induced apoptosis in the liver, which was confirmed by ultrastructure observation and changes in the expression of apoptosis indicators (P53, Bax, Bcl-2, Caspase-3, Caspase-9, Cleaved-Caspase-3 and Cytc). Additionally, co-exposure of As and PSNPs induced pyroptosis in the liver through NLRP3/Caspase-1 pathway via targeting NLRP3, ASC, Pro-Caspase-1, GSDMD and Cleaved-Caspase-1 expressions. Overall, our findings provide deeper insight into the roles of apoptosis, pyroptosis and excessive autophagy in the aggravation of liver injury, which could contribute to a better understanding of the interactions between As and PSNPS exposure and the molecular mechanisms of hepatotoxicity.


Assuntos
Arsênio , Doença Hepática Induzida por Substâncias e Drogas , Animais , Apoptose , Arsênio/toxicidade , Autofagia , Caspase 3 , Mamíferos/metabolismo , Camundongos , Microplásticos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Poliestirenos/metabolismo , Piroptose
9.
Toxicon ; 209: 18-27, 2022 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-35122786

RESUMO

Ducklings is one of the most susceptible poultry to Aflatoxin B1 (AFB1) which widely existed in duckling products will also in turn affect human health. Curcumin (CUR) has significant effects on immune regulation and anti-oxidation. But whether CUR alleviates toxic effects on duckling spleen induced by AFB1 remains largely unknown. In this study we treated duckings with AFB1 and CUR for 21 days before harvesting serum and spleen tissue for analyses. The results showed that AFB1 damaged the spleen tissue of ducklings by activating the NF-κB signaling pathway. And the addition of CUR not only promoted the growth of ducklings, but also enhanced the immune function of the spleen and reduced the damage of AFB1 to the spleen tissue. At the same time, CUR activated the Nrf2 signaling pathway, upregulated the expression of related antioxidant enzymes, inhibited the NF-kB signaling pathway, and ultimately reducing the inflammation of the duckling spleen induced by AFB1. It has been suggested from these results that Nrf2 pathway might be a potential therapeutic target for CUR to treat AFB1-induced immunosuppression in ducklings.


Assuntos
Aflatoxina B1 , Curcumina , Aflatoxina B1/metabolismo , Animais , Curcumina/farmacologia , Patos/metabolismo , Humanos , Terapia de Imunossupressão , Fator 2 Relacionado a NF-E2 , Baço/metabolismo
10.
Pancreatology ; 21(3): 630-641, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33707115

RESUMO

BACKGROUND: Pancreatic cancer (PC) is a highly fatal malignancy with few effective therapies currently available. Recent studies have shown that PD-L1 inhibitors could be potential therapeutic targets for the treatment of PC. The present study aims to investigate the effect of Shikonin on immune evasion in PC with the involvement of the PD-L1 degradation. METHODS: Initially, the expression patterns of PD-L1 and NF-κB in PC were predicted in-silico using the GEPIA database, and were subsequently validated using PC tissues. Thereafter, the correlation of NF-κB with STAT3, CSN5 and PD-L1 was examined. PC cells were treated with Shikonin, NF-κB inhibitor, STAT3 activator, and CSN5 overexpression plasmid to investigate effects on PD-L1 glycosylation and immune evasion in PC. Finally, in vivo tumor formation was induced in C57BL/6J mice, in order to verify the in vitro results. RESULTS: PD-L1, NF-κB, NF-κB p65, STAT3, and CSN5 were highly expressed in PC samples, and NF-κB was positively correlated with STAT3/CSN5/PD-L1. Inhibition of NF-κB decreased PD-L1 glycosylation and increased PD-L1 degradation, whereas activated STAT3 and overexpressed CSN5 reversed these trends. Shikonin blocked immune evasion in PC, and lowered the expression of PD-L1, NF-κB, NF-κB p65, STAT3 and CSN5 in vivo and in vitro. CONCLUSION: The findings indicated Shikonin inhibited immune evasion in PC by inhibiting PD-L1 glycosylation and activating the NF-κB/STAT3 and NF-κB/CSN5 signaling pathways. These effects of Shikonin on PC cells may bear important potential therapeutic implications for the treatment of PC.


Assuntos
Antineoplásicos/farmacologia , Antígeno B7-H1/metabolismo , Complexo do Signalossomo COP9/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , NF-kappa B/metabolismo , Naftoquinonas/farmacologia , Neoplasias Pancreáticas/tratamento farmacológico , Peptídeo Hidrolases/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/metabolismo , Western Blotting , Linhagem Celular Tumoral , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Naftoquinonas/uso terapêutico , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Evasão Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Transl Oncol ; 14(1): 100947, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33227664

RESUMO

Long non-coding RNAs (LncRNAs) can regulate physiological and pathological functions, exhibiting a wide range of roles in cell biology. Moreover, many lncRNAs are dysregulated in various cancers, including colon cancer. In this study, we investigated the role of the lncRNA LINC00355 in colon cancer, after first establishing its interaction with GTF2B, and ITGA2 on the LncMap database. The predicted relationships between the lncRNA LINC00355, GTF2B, and ITGA2 were identified using luciferase reporter assay, RIP, and ChIP experiments. Western blot analysis and RT-qPCR were applied to determine expression pattern of lncRNA LINC00355 and ITGA2 in colon cancer cells. Additionally, EdU, TUNEL, Cell-adhesion and Transwell assay was used for the detection of the effects of this axis on proliferation, apoptosis, adhesion, chemotaxis and metastasis. LncRNA LINC00355 targeted IGFBP2 through the recruitment of GTF2B. LncRNA LINC00355 was highly expressed in colon cancer cells, and overexpression of lncRNA LINC00355 increased the expression of IGFBP2 and GTF2B, and thereby promoted the proliferation, chemotaxis, invasion, and migration in colon cancer. In summary, downregulation of lncRNA LINC00355 in colon cancer inhibited tumor growth in colon cancer through effects on the GTF2B/IGFBP2 axis.

12.
Cancer Cell Int ; 20: 511, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33093810

RESUMO

BACKGROUND: Colorectal cancer (CRC) is considered as the second common death-induced cancer. More recently, association of long non-coding RNAs (lncRNAs) with CRC has been extensively investigated. Therefore, the present study was performed to determine whether lncRNA MAF BZIP Transcription Factor G Antisense RNA 1 (MAFG-AS1) could regulate biological activities of CRC cells and unravel the underlying mechanisms. METHODS: CRC and corresponding adjacent tissues were collected to determine the expression of lncRNA MAFG-AS1, microRNA-149-3p (miR-149-3p) and homeobox B8 (HOXB8) by RT-qPCR. Dual luciferase reporter gene assay was used to explore the targeting relationship between miR-149-3p and lncRNA MAFG-AS1 and between miR-149-3p and HOXB8, followed by RNA immunoprecipitation for verification. Migration, proliferation, invasion, and apoptosis of HCT116 and LoVo cells were examined when lncRNA MAFG-AS1 was silenced or miR-149-3p was overexpressed. Furthermore, tumorigenicity of HCT116 and LoVo cells was measured in vivo by tumor xenograft in nude mice. RESULTS: LncRNA MAFG-AS1 and HOXB8 were found to be highly expressed in CRC tissues and cells, while miR-149-3p was under-expressed. LncRNA MAFG-AS1 negatively regulated miR-149-3p while miR-149-3p downregulated HOXB8. In addition, lncRNA MAFG-AS1 silencing by shRNA or miR-149-3p upregulation by mimic suppressed the migration, proliferation, invasion and tumorigenesis but promoted the apoptosis of HCT116 and LoVo cells. CONCLUSION: Taken together, lncRNA MAFG-AS1 downregulation inhibits the malignant behaviors of CRC cells by upregulating miR-149-3p and downregulating HOXB8, providing a potential therapeutic target for CRC treatment.

13.
Cell Biol Int ; 44(1): 306-316, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31483550

RESUMO

Fatty liver disease is a disease manifested with excessive alcohol intake and obese. Importantly, hydrogen sulfide (H2 S) has been revealed to participate in the progression of fatty liver; however, the underlying mechanism has not been clearly elucidated yet. In this study, we aimed to investigate the effects of exogenous H2 S on fatty liver ischemia-reperfusion injury (IRI) through mediating class A scavenger receptor (SRA) pathway in rats. By determining endoplasmic reticulum stress (ERS)-related factors, autophagy markers and apoptosis-related factors in liver tissue and liver function, levels of oxidative stress, inflammatory factors, and hepatocyte apoptosis, the effects of H2 S on IRI-induced autophagy, oxidative stress, and inflammation were all examined in rat model of fatty liver IRI. Results from obtained data showed that H2 S decreased the expression of SRA, Grp78, PERK, CHOP, and Caspase-3, and increased that of LC3-II/LC3-I, in addition to alleviating the pathological changes of liver and reducing the levels of ALT, AST, LDH TBARS, and MDA. Moreover, H2 S decreased the levels of oxidative stress, the expression of pro-inflammatory factors including tumor necrosis factor α, interleukin 1, and interleukin 6, and the apoptosis of hepatocytes. Our findings suggested exogenous H2 S could reduce ERS by mediating the SRA pathway and protect liver function by inducing autophagy, and protect against IRI by reducing oxidative stress and inflammation.

14.
Int Immunopharmacol ; 78: 106003, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31812723

RESUMO

Cancer stem cells (CSCs) are considered to mediate tumorigenesis, recurrence, and metastasis. KYA1797K, a ß-catenin inhibitor, has been identified for its functionality as a tumor suppressor gene in colorectal cancer through inhibition of the Wnt/ß-catenin signaling pathway. However, it remains uncertain whether KYA1797K attenuates immune evasion of colon CSCs. Hence, this study is designed for evaluating the function of KYA1797K in colon CSCs. The expression of ß-catenin and STT3A/B in colon cancer tissues was initially detected by immunohistochemistry, followed by correlation analyses of the survival rate with the expression of ß-catenin and STT3A/B as well as identification of the interaction between ß-catenin and STT3A/B. Besides, ß-catenin in colon CSCs was knocked down or inhibited by KYA1797K to explore its role in immune evasion and the subsequent underlying mechanism associated with STT3A/B expression and PD-L1 glycosylation. Additionally, the in vivo regulatory effects of ß-catenin silencing and KYA1797K were evaluated by assessing tumor formation, detecting CD8 and GZMB expression and CD8+ T cell viability. The results collected displayed that ß-catenin and STT3A/B showed high expression in colon cancer tissues, both of which were correlated with poor survival of colon cancer patients. ß-catenin was found to positively regulate STT3A/B expression. Besides, ß-catenin silencing or KYA1797K treatment down-regulated the expression of STT3A/B, inhibited PD-L1 glycosylation and suppressed immune evasion of colon CSCs both in vivo and in vitro. Altogether, KYA1797K inhibits the ß-catenin/STT3 signaling pathway to reduce the stability of PD-L1, thus further inhibiting immune evasion and inducing apoptosis of colon CSCs, which contributes to the development of immunotherapy for colon cancer.


Assuntos
Neoplasias do Colo/imunologia , Evasão da Resposta Imune/efeitos dos fármacos , Células-Tronco Neoplásicas/efeitos dos fármacos , Tiazolidinas/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos , Animais , Antígeno B7-H1/imunologia , Linhagem Celular , Colo/imunologia , Regulação para Baixo , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células-Tronco Neoplásicas/imunologia
15.
Int J Mol Sci ; 20(10)2019 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-31137617

RESUMO

Liver regeneration plays a significant role in protecting liver function after liver injury or chronic liver disease. Long non-coding RNAs (lncRNAs) are considered to be involved in the proliferation of hepatocytes and liver regeneration. Therefore, this study aimed to explore the effects of LncRNA-Dreh on the regulation of hepatic progenitor cells (HPCs) during liver regeneration in rats. Initially, the rat model of liver injury was established to investigate the effect of LncRNA-Dreh down-regulation on liver tissues of rats with liver injury. Subsequently, HPCs line WB-F344 cells were transfected with interference plasmid of LncRNA-Dreh and the expression of LncRNA-Dreh and Vimentin was detected. The proliferation and migration ability of WB-F344 cells, as well as the content of albumin (ALB) and alpha fetoprotein (AFP) in cell differentiation were then determined. Disorderly arranged structure of liver tissue, a large number of HPCs set portal area as center extended to hepatic lobule and ductular reaction were observed in liver tissues of rats with liver injury. The expression of LncRNA-Dreh decreased while Vimentin increased in liver tissues of rats with liver injury. Moreover, the proliferation and migration ability, expression of Vimentin and AFP in WB-F344 cells were increased after silencing of LncRNA-Dreh and ALB was decreased. Collectively, our findings demonstrate that inhibition of LncRNA-Dreh can enhance the proliferation and migration abilities of HPCs in liver regeneration but cause abnormal differentiation of HPCs.


Assuntos
Movimento Celular , Proliferação de Células , Hepatócitos/metabolismo , Regeneração Hepática , RNA Longo não Codificante/genética , Células-Tronco/metabolismo , Animais , Linhagem Celular , Hepatócitos/fisiologia , Masculino , RNA Longo não Codificante/metabolismo , Ratos , Ratos Sprague-Dawley , Células-Tronco/fisiologia , Vimentina/genética , Vimentina/metabolismo
16.
Parasitol Res ; 110(2): 961-9, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21842385

RESUMO

T lymphocytes play a vital role in antimalaria immunity, but there is little information about the role of T cells in malaria infection. In order to explore the profile of T cells in malaria immunity, we infected Chinese rhesus macaques with the malaria parasite (Plasmodium cynomolgi) and examined the dynamics of T cell subsets. Both repeated and long-term infections were involved. Our results showed that the monkeys in the repeated infection group acquired protective immunity through primary infection, which was evidenced by a much lower parasitemia, milder anemia, and milder fever during reinfection; the monkeys in the long-term infection group also developed protective immunity, but this was not sufficient to eliminate the parasite. The total counts of leukocytes, neutrophils, CD3+ T cells, CD4+ or CD8+ T cells, and naïve and memory CD4+ and CD8+ T cells declined during the acute phase of malaria but increased after the parasite was controlled. The total number of activated CD4+ T cells significantly increased during malaria in animals with a long-term infection, which remained at least 3 months after the termination of malaria. However, the activated CD4+ T cells decreased during the acute phase of infection in the repeated infection group and converted to preinfection levels after malaria was cured. Regulatory CD4+ T cells continued to increase during the malaria infections and quickly reverted to preinfection levels after the parasite was controlled. Our study provides a systematic analysis of the kinetic profiles of T lymphocyte subsets during malaria infections and provides some experimental insight into malaria immunology.


Assuntos
Sangue/imunologia , Macaca mulatta/imunologia , Malária/veterinária , Plasmodium cynomolgi/imunologia , Doenças dos Primatas/imunologia , Subpopulações de Linfócitos T/imunologia , Anemia/prevenção & controle , Animais , Complexo CD3/análise , Antígenos CD4/análise , Antígenos CD8/análise , China , Modelos Animais de Doenças , Imunofenotipagem , Malária/imunologia , Malária/parasitologia , Malária/patologia , Parasitemia/prevenção & controle , Doenças dos Primatas/parasitologia , Doenças dos Primatas/patologia , Subpopulações de Linfócitos T/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA