Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Front Med (Lausanne) ; 11: 1368977, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38947241

RESUMEN

Intestinal fibrosis is a common complication of chronic intestinal diseases with the characteristics of fibroblast proliferation and extracellular matrix deposition after chronic inflammation, leading to lumen narrowing, structural and functional damage to the intestines, and life inconvenience for the patients. However, anti-inflammatory drugs are currently generally not effective in overcoming intestinal fibrosis making surgery the main treatment method. The development of intestinal fibrosis is a slow process and its onset may be the result of the combined action of inflammatory cells, local cytokines, and intestinal stromal cells. The aim of this study is to elucidate the pathogenesis [e.g., extracellular matrix (ECM), cytokines and chemokines, epithelial-mesenchymal transition (EMT), differentiation of fibroblast to myofibroblast and intestinal microbiota] underlying the development of intestinal fibrosis and to explore therapeutic advances (such as regulating ECM, cytokines, chemokines, EMT, differentiation of fibroblast to myofibroblast and targeting TGF-ß) based on the pathogenesis in order to gain new insights into the prevention and treatment of intestinal fibrosis.

2.
Biochem Biophys Res Commun ; 728: 150340, 2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-38968770

RESUMEN

BACKGROUND: Colorectal cancer (CRC) is a prevalent malignancy and poses a significant clinical challenge. Piperine, an alkaloid molecule extracted from Piper nigrum and Piper longum, has emerged as a promising anticancer agent. However, the molecular mechanisms of piperine' antitumor effects in CRC need to be further elucidated. METHODS: Human colorectal cancer cells were treated with piperine in vitro. CCK-8 and clone formation assays were adopted to detect cell viability. The accumulation of autophagosomes was assessed by Western blotting and immunofluorescence. Apoptosis and reactive oxygen species (ROS) levels were analyzed by flow. In vivo, a xenograft tumor mouse model was constructed using CT26 cells. RESULTS: Piperine inhibited CRC cell viability and suppressed tumor weight and volume in a mouse model. Additionally, piperine treatment induced the accumulation of autophagosomes in CRC cells. This effect was attributed to the inhibition of the AKT/mTOR pathway and the accumulation of ROS. activation of AKT or clearance of ROS attenuated piperine-mediated tumor suppression. CONCLUSION: This study shows that piperine induces autophagy-dependent cell death in CRC cells by increasing ROS production and inhibiting Akt/mTOR signaling.


Asunto(s)
Alcaloides , Autofagia , Benzodioxoles , Neoplasias del Colon , Piperidinas , Alcamidas Poliinsaturadas , Proteínas Proto-Oncogénicas c-akt , Especies Reactivas de Oxígeno , Transducción de Señal , Serina-Treonina Quinasas TOR , Alcamidas Poliinsaturadas/farmacología , Benzodioxoles/farmacología , Piperidinas/farmacología , Alcaloides/farmacología , Especies Reactivas de Oxígeno/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Humanos , Serina-Treonina Quinasas TOR/metabolismo , Autofagia/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Neoplasias del Colon/metabolismo , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Línea Celular Tumoral , Ratones , Ratones Endogámicos BALB C , Ensayos Antitumor por Modelo de Xenoinjerto , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ratones Desnudos
3.
Med Oncol ; 39(12): 211, 2022 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-36175702

RESUMEN

Colorectal cancer (CRC) is a cancer with a high morbidity and mortality worldwide. Hence, developing new therapeutic drugs for CRC is very important. Moxidectin (MOX) has shown good anti-glioblastoma effect both in vitro and in vivo. This study aimed to elucidate the anti-CRC effect of MOX and its potential mechanism by investigating the influence of MOX on the viability, apoptosis, necrosis and autophagy of colorectal cancer cells (HCT15 and SW620) and its underlying mechanisms. It was found that MOX can induce autophagy arrest, promote autophagy initiation, inhibit autophagic flux and cell proliferation, simultaneously PI3K-Akt-mTOR signaling pathway and microtubule acetylation. Furthermore, MOX suppressed the growth of xenograft tumors, which was consistent with the in vitro results.


Asunto(s)
Neoplasias Colorrectales , Fosfatidilinositol 3-Quinasas , Autofagia , Neoplasias Colorrectales/tratamiento farmacológico , Humanos , Macrólidos , Proteínas Proto-Oncogénicas c-akt , Serina-Treonina Quinasas TOR
4.
Theranostics ; 12(10): 4802-4817, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35832094

RESUMEN

Background: Chemoresistance to 5-fluorouracil (5-FU) is a major barrier to influence the treatment efficiency of colorectal cancer (CRC) patients, while the precise molecular mechanisms underlying 5-FU resistance remain to be fully elucidated. Methods: The metabolic profiles including ATP generation, glucose consumption, lactate generation, and oxygen consumption rate (OCR) in 5-FU resistant CRC cells were compared with those in their parental cells. Subsequently, a series of in vitro and in vivo experiments were carried out to investigate the mechanisms responsible for metabolic reprogramming of 5-FU resistant CRC cells. Results: We found that 5-FU resistant CRC cells showed increased levels of ATP generation, glucose consumption, lactate generation, and OCR as compared with those in their parental cells. Further, increased levels of mRNA N6-methyladenosine (m6A) and methyltransferase-like 3 (METTL3) were observed in 5-FU resistant CRC cells. Inhibition or knockdown of METTL3 can suppress glycolysis and restore chemosensitivity of 5-FU resistant CRC cells. Mechanistically, METTL3 enhances the expression of LDHA, which catalyzes the conversion of pyruvate to lactate, to trigger glycolysis and 5-FU resistance. METTL3 can increase the transcription of LDHA via stabilizing mRNA of hypoxia-inducible factor (HIF-1α), further, METTL3 also triggers the translation of LDHA mRNA via methylation of its CDS region and recruitment of YTH domain-containing family protein 1 (YTHDF1). Targeted inhibition of METTL3/LDHA axis can significantly increase the in vitro and in vivo 5-FU sensitivity of CRC cells. Conclusion: Our study indicates that METTL3/LDHA axis-induced glucose metabolism is a potential therapy target to overcome 5-FU resistance in CRC cells.


Asunto(s)
Adenosina , Neoplasias Colorrectales , Fluorouracilo , L-Lactato Deshidrogenasa , Adenosina/análogos & derivados , Adenosina Trifosfato/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/metabolismo , Resistencia a Antineoplásicos , Fluorouracilo/farmacología , Glucosa/metabolismo , Células HCT116 , Humanos , L-Lactato Deshidrogenasa/biosíntesis , L-Lactato Deshidrogenasa/genética , L-Lactato Deshidrogenasa/metabolismo , Lactatos/metabolismo , Metiltransferasas/genética , ARN Mensajero
5.
Int J Biol Sci ; 18(6): 2540-2552, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35414768

RESUMEN

Colorectal cancer (CRC) is one of the most common malignancies worldwide. Metastasis is a major cause of CRC recurrence and mortality. Several antibiotic drugs have been reported to exert potential anticancer activities, however, whether and how the tetracycline antibiotic minocycline exhibit tumor suppressive effect on CRC remains unknown. Here, we found that minocycline markedly inhibits the epithelial-mesenchymal transition (EMT) process and metastasis of CRC cells both in vitro and in vivo. Using chemical proteomics screening combined with docking analysis and site-directed mutagenesis, we identified LYN as a direct bind target of minocycline, and Ala255 of LYN is required for minocycline binding. Mechanistically, minocycline binding inactivates LYN, leading to STAT3 inactivation and EMT suppression, thereby inhibits CRC metastasis. Tissue microarray analysis further confirmed the clinical relevance of LYN-STAT3 axis in the EMT and progression of CRC. In addition to CRC, minocycline also significantly prevents EMT process and inhibits the metastasis of several other cancer types. Our findings elucidate the mechanism of action of minocycline for the inhibition of CRC metastasis by LYN binding, and suggest that repurposing minocycline may represent a promising strategy for the treatment of advanced CRC and other cancer types.


Asunto(s)
Neoplasias Colorrectales , Minociclina , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Línea Celular Tumoral , Movimiento Celular , Neoplasias Colorrectales/metabolismo , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Humanos , Minociclina/farmacología , Minociclina/uso terapéutico , Metástasis de la Neoplasia , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo
6.
Aging (Albany NY) ; 12(23): 24424-24440, 2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33291076

RESUMEN

Colorectal cancer (CRC) is the third most common type of cancer worldwide. Metastasis and chemoresistance are regarded as the two leading causes of treatment failure and high mortality in CRC. Forkhead Box M1 (FOXM1) has been involved in malignant behaviors of cancer. However, the role and mechanism of FOXM1 in simultaneously regulating metastasis and chemoresistance of CRC remain poorly understood. Here, we found that FOXM1 was overexpressed in oxaliplatin- and vincristine-resistant CRC cells (HCT-8/L-OHP and HCT-8/VCR) with enhanced metastatic potential, compared with HCT-8 cells. FOXM1 overexpression increased migration, invasion and drug-resistance to oxaliplatin and vincristine in HCT-8 cells, while FOXM1 knockdown using shFOXM1 impaired metastasis and drug-resistance in HCT-8/L-OHP and HCT-8/VCR cells. Moreover, FOXM1 up-regulated Snail to trigger epithelial-mesenchymal transition-like molecular changes and multidrug-resistance protein P-gp expression, while silencing Snail inhibited FOXM1-induced metastasis and drug-resistance. We further identified that disheveled-2 (DVL2) was crucial for FOXM1-induced Snail expression, metastasis and chemoresistance. Furthermore, FOXM1 bound to DVL2, and enhanced nuclear translocation of DVL2 and DVL2-mediated transcriptional activity of Wnt/ß-catenin known to induce Snail expression. In conclusion, FOXM1/DVL2/Snail axis triggered aggressiveness of CRC. Blocking FOXM1/DVL2/Snail pathway simultaneously inhibited metastasis and chemoresistance in CRC cells, providing a new strategy for successful CRC treatment.


Asunto(s)
Antineoplásicos/farmacología , Movimiento Celular/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Proteínas Dishevelled/metabolismo , Resistencia a Antineoplásicos , Proteína Forkhead Box M1/metabolismo , Oxaliplatino/farmacología , Factores de Transcripción de la Familia Snail/metabolismo , Vincristina/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Proteínas Dishevelled/genética , Resistencia a Antineoplásicos/genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Proteína Forkhead Box M1/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Metástasis de la Neoplasia , Factores de Transcripción de la Familia Snail/genética , Vía de Señalización Wnt
7.
Cancer Cell Int ; 20: 205, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32514250

RESUMEN

BACKGROUND: Inflammation is one of a main reason for colon cancer progression and poor prognosis. The high-mobility group box-1 (HMGB1) and glutathione peroxidase 4 (GPX4) are responsible for inflammation, but the relationship between HMGB1 and GPX4 remains unknown about inflammation in colon cancer. METHODS: RT-qPCR was carried out to investigate the expression of IL1ß, IL6 and TNFα in colon cancer cells stimulated with LPS or siHMGB1. To observe the relationship between HMGB1, GPX4 and inflammation or ROS, Western blot assays were adopted. Pull-down, CoIP and immunohistochemistry assays were performed to further investigate the molecular mechanisms of HMGB1 and GPX4 in colon cancer. RESULTS: We report that HMGB1 mediates lipopolysaccharide (LPS)-induced inflammation in colon cancer cells. Mechanistically, acetylated HMGB1 interacts with GPX4, negatively regulating GPX4 activity. Furthermore, by utilizing siHMGB1 and its inhibitor, our discoveries demonstrate that HMGB1 knockdown can inhibit inflammation and reactive oxygen species (ROS) accumulation via NF-kB. CONCLUSION: Collectively, our findings first demonstrate that acetylated HMGB1 can interact with GPX4, leading to inflammation, and providing therapeutic strategies targeting HMGB1 and GPX4 for colon cancer.

8.
Cell Prolif ; 51(3): e12427, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29292543

RESUMEN

OBJECTIVES: Alzheimer's disease (AD) is one of the most prevalent brain diseases among the elderly, majority of which is caused by abnormal deposition of amyloid beta-peptide (Aß). Galantamine, currently the first-line drug in treatment of AD, has been shown to diminish Aß-induced neurotoxicity and exert favourable neuroprotective effects, but the detail mechanisms remain unclear. MATERIALS AND METHODS: Effects of galantamine on Aß-induced cytotoxicity were checked by MTT, clone formation and apoptosis assays. The protein variations and reactive oxygen species (ROS) production were measured by western blotting analysis and dichloro-dihydro-fluorescein diacetate assay, respectively. RESULTS: Galantamine reversed Aß-induced cell growth inhibition and apoptosis in neuron cells PC12. Aß activated the entire autophagy flux and accumulation of autophagosomes, and the inhibition of autophagy decreased the protein level of cleaved-caspase-3 and Aß-induced cytotoxicity. Meanwhile, galantamine suppressed Aß-mediated autophagy flux and accumulation of autophagosomes. Moreover, Aß upregulated ROS accumulation, while ROS scavengers N-acetyl-l-cysteine impaired Aß-mediated autophagy. Further investigation showed that galantamine downregulated NOX4 expression to inhibit Aß-mediated ROS accumulation and autophagy. CONCLUSIONS: Galantamine inhibits Aß-induced cytostatic autophagy through decreasing ROS accumulation, providing new insights into deep understanding of AD progression and molecular basis of galantamine in neuroprotection.


Asunto(s)
Péptidos beta-Amiloides/fisiología , Autofagia/efectos de los fármacos , Citostáticos/farmacología , Galantamina/farmacología , Fragmentos de Péptidos/fisiología , Especies Reactivas de Oxígeno/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Animales , Apoptosis , Proliferación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , NADPH Oxidasa 4/metabolismo , Células PC12 , Ratas
9.
J Cell Mol Med ; 22(1): 89-100, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28799229

RESUMEN

Recently, the dipeptidyl peptidase-4 (DPP-4) inhibitor sitagliptin, a major anti-hyperglycaemic agent, has received substantial attention as a therapeutic target for cardiovascular diseases via enhancing the number of circulating endothelial progenitor cells (EPCs). However, the direct effects of sitagliptin on EPC function remain elusive. In this study, we evaluated the proangiogenic effects of sitagliptin on a diabetic hind limb ischaemia (HLI) model in vivo and on EPC culture in vitro. Treatment of db/db mice with sitagliptin (Januvia) after HLI surgery efficiently enhanced ischaemic angiogenesis and blood perfusion, which was accompanied by significant increases in circulating EPC numbers. EPCs derived from the bone marrow of normal mice were treated with high glucose to mimic diabetic hyperglycaemia. We found that high glucose treatment induced EPC apoptosis and tube formation impairment, which were significantly prevented by sitagliptin pretreatment. A mechanistic study found that high glucose treatment of EPCs induced dramatic increases in oxidative stress and apoptosis; pretreatment of EPCs with sitagliptin significantly attenuated high glucose-induced apoptosis, tube formation impairment and oxidative stress. Furthermore, we found that sitagliptin restored the basal autophagy of EPCs that was impaired by high glucose via activating the AMP-activated protein kinase/unc-51-like autophagy activating kinase 1 signalling pathway, although an autophagy inhibitor abolished the protective effects of sitagliptin on EPCs. Altogether, the results indicate that sitagliptin-induced preservation of EPC angiogenic function results in an improvement of diabetic ischaemia angiogenesis and blood perfusion, which are most likely mediated by sitagliptin-induced prevention of EPC apoptosis via augmenting autophagy.


Asunto(s)
Autofagia , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Células Progenitoras Endoteliales/patología , Isquemia/tratamiento farmacológico , Neovascularización Fisiológica , Fosfato de Sitagliptina/uso terapéutico , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Autofagia/efectos de los fármacos , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/patología , Supervivencia Celular/efectos de los fármacos , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/patología , Células Progenitoras Endoteliales/efectos de los fármacos , Células Progenitoras Endoteliales/metabolismo , Glucosa/toxicidad , Miembro Posterior/irrigación sanguínea , Isquemia/complicaciones , Isquemia/patología , Masculino , Ratones , Neovascularización Fisiológica/efectos de los fármacos , Perfusión , Especies Reactivas de Oxígeno/metabolismo , Flujo Sanguíneo Regional/efectos de los fármacos , Transducción de Señal , Fosfato de Sitagliptina/farmacología
10.
Circ Res ; 120(5): e7-e23, 2017 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-28137917

RESUMEN

RATIONALE: Endothelial progenitor cells (EPCs) respond to stromal cell-derived factor 1 (SDF-1) through chemokine receptors CXCR7 and CXCR4. Whether SDF-1 receptors involves in diabetes mellitus-induced EPCs dysfunction remains unknown. OBJECTIVE: To determine the role of SDF-1 receptors in diabetic EPCs dysfunction. METHODS AND RESULTS: CXCR7 expression, but not CXCR4 was reduced in EPCs from db/db mice, which coincided with impaired tube formation. Knockdown of CXCR7 impaired tube formation of EPCs from normal mice, whereas upregulation of CXCR7 rescued angiogenic function of EPCs from db/db mice. In normal EPCs treated with oxidized low-density lipoprotein or high glucose also reduced CXCR7 expression, impaired tube formation, and increased oxidative stress and apoptosis. The damaging effects of oxidized low-density lipoprotein or high glucose were markedly reduced by SDF-1 pretreatment in EPCs transduced with CXCR7 lentivirus but not in EPCs transduced with control lentivirus. Most importantly, EPCs transduced with CXCR7 lentivirus were superior to EPCs transduced with control lentivirus for therapy of ischemic limbs in db/db mice. Mechanistic studies demonstrated that oxidized low-density lipoprotein or high glucose inhibited protein kinase B and glycogen synthase kinase-3ß phosphorylation, nuclear export of Fyn and nuclear localization of nuclear factor (erythroid-derived 2)-like 2 (Nrf2), blunting Nrf2 downstream target genes heme oxygenase-1, NAD(P)H dehydrogenase (quinone 1) and catalase, and inducing an increase in EPC oxidative stress. This destructive cascade was blocked by SDF-1 treatment in EPCs transduced with CXCR7 lentivirus. Furthermore, inhibition of phosphatidylinositol 3-kinase/protein kinase B prevented SDF-1/CXCR7-mediated Nrf2 activation and blocked angiogenic repair. Moreover, Nrf2 knockdown almost completely abolished the protective effects of SDF-1/CXCR7 on EPC function in vitro and in vivo. CONCLUSIONS: Elevated expression of CXCR7 enhances EPC resistance to diabetes mellitus-induced oxidative damage and improves therapeutic efficacy of EPCs in treating diabetic limb ischemia. The benefits of CXCR7 are mediated predominantly by a protein kinase B/glycogen synthase kinase-3ß/Fyn pathway via increased activity of Nrf2.


Asunto(s)
Diabetes Mellitus/metabolismo , Células Progenitoras Endoteliales/fisiología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Isquemia/metabolismo , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Receptores CXCR/biosíntesis , Animales , Células Cultivadas , Diabetes Mellitus/patología , Técnicas de Silenciamiento del Gen , Células HEK293 , Miembro Posterior/irrigación sanguínea , Miembro Posterior/metabolismo , Miembro Posterior/patología , Humanos , Isquemia/patología , Masculino , Ratones , Ratones Transgénicos , Factor 2 Relacionado con NF-E2/metabolismo , Neovascularización Fisiológica/fisiología , Estrés Oxidativo/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA