Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 597
Filtrar
1.
Ecotoxicol Environ Saf ; 268: 115689, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37992645

RESUMEN

Avian tibial dyschondroplasia (TD) is a skeletal disease affecting fast growing chickens, resulting in non-mineralized avascular cartilage. This metabolic disorder is characterized by lameness and reduced growth performance causing economic losses. The aim of this study was to investigate the protective effects of baicalin against TD caused by thiram exposure. A total of two hundred and forty (n = 240) one day-old broiler chickens were uniformly and randomly allocated into three different groups (n = 80) viz. control, TD, and baicalin groups. All chickens received standard feed, however, to induce TD, the TD and baicalin groups received thiram (tetramethylthiuram disulfide) at a rate of 50 mg/kg feed from days 4-7. The thiram induction in TD and baicalin groups resulted in lameness, high mortality, and enlarged growth-plate, poor production performance, reduction in ALP, GSH-Px, SOD, and T-AOC levels, and increased AST and ALT, and MDA levels. Furthermore, histopathological results showed less vascularization, and mRNA and protein expression levels of Sox-9, Col-II, and Bcl-2 showed significant downward trend, while caspase-9 displayed significant up-regulation in TD-affected chickens. After the TD induction, the baicalin group was orally administered with baicalin at a rate of 200 mg/kg from days 8-18. Baicalin administration increased the vascularization, and chondrocytes with intact nuclei, alleviated lameness, decreased GP size, increased productive capacity, and restored the liver antioxidant enzymes and serum biochemical levels. Furthermore, baicalin significantly up-regulated the gene and protein expressions of Sox-9, Col-II, and Bcl-2, and significantly down-regulated the expression of caspase-9 (p < 0.05). Therefore, the obtained results suggest that baicalin could be a possible choice in thiram toxicity alleviation by regulating apoptosis and chondrocyte proliferation in thiram-induced tibial dyschondroplasia.


Asunto(s)
Osteocondrodisplasias , Tiram , Animales , Tiram/toxicidad , Osteocondrodisplasias/inducido químicamente , Osteocondrodisplasias/genética , Pollos , Condrocitos/patología , Caspasa 9/genética , Cojera Animal , Apoptosis , Neovascularización Patológica/inducido químicamente , Proliferación Celular
2.
Ecotoxicol Environ Saf ; 239: 113674, 2022 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-35623148

RESUMEN

N-methyl-N-nitrosourea (MNU) is a prevalent environmental carcinogen, which leads to tumors in various organs in animal models, while the mechanisms involved were still not fully understood. It is well known that anomalous angiogenesis is a key step in tumorigenesis and progression. In this study, we found that MNU induced abnormal angiogenesis which was accompanied by upregulation of rspo1, p53 and vegfaa in zebrafish embryos. Moreover, it revealed that MNU-induced ectopic sprouting of blood vessels was significantly reduced in rspo1-knockdown but not p53-knockdown embryos, indicating that rspo1 was necessary for MNU-induced abnormal angiogenesis. Additionally, pharmaceutical activation or inhibition of Wnt/ß-catenin signaling pathway using (2'Z,3'E)- 6-bromoindirubin-3'-oxime or CCT036477 significantly increased or inhibited the pro-angiogenic effect of MNU on developing zebrafish embryos, which was confirmed by the effect of proliferation and migration in MNU-treated bEnd.3 cells. These data together indicated that rspo1/Wnt/ß-catenin/vegfaa axis is involved in the modulation of MNU-induced anomalous angiogenesis.


Asunto(s)
Metilnitrosourea , Neovascularización Patológica , Vía de Señalización Wnt , Pez Cebra , Animales , Células Endoteliales/metabolismo , Metilnitrosourea/toxicidad , Ratones , Neovascularización Patológica/inducido químicamente , Pez Cebra/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
3.
Int J Mol Sci ; 23(3)2022 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-35163148

RESUMEN

Increased soluble endoglin (sENG) has been observed in human brain arteriovenous malformations (bAVMs). In addition, the overexpression of sENG in concurrence with vascular endothelial growth factor (VEGF)-A has been shown to induce dysplastic vessel formation in mouse brains. However, the underlying mechanism of sENG-induced vascular malformations is not clear. The evidence suggests the role of sENG as a pro-inflammatory modulator, and increased microglial accumulation and inflammation have been observed in bAVMs. Therefore, we hypothesized that microglia mediate sENG-induced inflammation and endothelial cell (EC) dysfunction in bAVMs. In this study, we confirmed that the presence of sENG along with VEGF-A overexpression induced dysplastic vessel formation. Remarkably, we observed increased microglial activation around dysplastic vessels with the expression of NLRP3, an inflammasome marker. We found that sENG increased the gene expression of VEGF-A, pro-inflammatory cytokines/inflammasome mediators (TNF-α, IL-6, NLRP3, ASC, Caspase-1, and IL-1ß), and proteolytic enzyme (MMP-9) in BV2 microglia. The conditioned media from sENG-treated BV2 (BV2-sENG-CM) significantly increased levels of angiogenic factors (Notch-1 and TGFß) and pERK1/2 in ECs but it decreased the level of IL-17RD, an anti-angiogenic mediator. Finally, the BV2-sENG-CM significantly increased EC migration and tube formation. Together, our study demonstrates that sENG provokes microglia to express angiogenic/inflammatory molecules which may be involved in EC dysfunction. Our study corroborates the contribution of microglia to the pathology of sENG-associated vascular malformations.


Asunto(s)
Endoglina/administración & dosificación , Endotelio Vascular/patología , Inflamación/patología , Microglía/patología , Neovascularización Patológica/patología , Enfermedades Vasculares/patología , Factor A de Crecimiento Endotelial Vascular/administración & dosificación , Animales , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Inflamación/inducido químicamente , Inflamación/inmunología , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/inmunología , Microglía/metabolismo , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/inmunología , Neovascularización Patológica/metabolismo , Enfermedades Vasculares/inducido químicamente , Enfermedades Vasculares/inmunología , Enfermedades Vasculares/metabolismo , Factor A de Crecimiento Endotelial Vascular/efectos adversos
4.
Toxicol Appl Pharmacol ; 436: 115854, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34974051

RESUMEN

BACKGROUND: Colorectal cancer (CRC) is one of the most malignant cancers worldwide. Nonylphenol (NP) is an endocrine-disruptor chemical and plays an important role in the development of cancers. However, the effects of NP on CRC remain unclear. In this study, we aimed to investigate the potential mechanisms of NP in the pathogenesis of CRC. METHODS: The levels of AhR, TL1A and HDAC2 in CRC tissues and endothelial cells were assessed by RT-qPCR or western blot. CHIP and dual luciferase reporter assays were used to confirm the interaction between AhR and HDAC2, or HNF4α and TL1A. The CCK8, would healing and tube formation assays were conducted to evaluate the proliferation, migration and angiogenesis of HUVECs. Western blot determined HNF4α protein and HNF4α acetylation levels. The secreted TL1A protein was detected by ELISA. The angiogenesis-related factor CD31 was tested by IHC. RESULTS: The expression level of AhR was significantly up-regulated in CRC tissues and endothelial cells. Moreover, NP activated the AhR pathway mediated colorectal endothelial cell angiogenesis and proliferation, while TL1A overexpression resisted these effects caused by NP. Besides, NP was found to modulate HNF4α deacetylation through AhR/HDAC2 to inhibit TL1A. Furthermore, in vivo experiments proved that NP regulated CRC growth and angiogenesis via AhR/HDAC2/HNF4α/TL1A axis. CONCLUSION: This study revealed that NP promoted CRC growth and angiogenesis through AhR/HDAC2/HNF4α/TL1A pathway and could be a new therapeutic target for CRC treatment.


Asunto(s)
Neoplasias Colorrectales/inducido químicamente , Factor Nuclear 4 del Hepatocito/metabolismo , Histona Desacetilasa 2/metabolismo , Neovascularización Patológica/inducido químicamente , Fenoles/efectos adversos , Receptores de Hidrocarburo de Aril/metabolismo , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Células Cultivadas , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
5.
Drug Chem Toxicol ; 45(2): 589-593, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32126859

RESUMEN

Lead (Pb) is a toxic heavy metal ubiquitously distributed around the world, especially in industrial areas. Occupational and environmental exposures to Pb have detrimental effects on human health. Pb affects functioning of many systems of the human body, including the cardiovascular system. Angiogenesis, the process of new blood vessel formation, which makes critical contribution throughout life is deranged in various diseases. Excessive angiogenesis may result in different diseases including cancer. On the other spectrum, insufficient angiogenesis is observed in many diseases, such as atherosclerosis, hypertension, and cardiovascular disease. These disorders are also associated with occupational Pb exposure. In this paper, epidemiological and experimental studies are reviewed selectively for evidence in support of this hypothesis, that is, interactions between Pb and angiogenesis. We discuss the evidence for the possible mechanism of Pb impact on concentrations of angiogenic factors. Studies suggested that Pb exposure affects the level of angiogenic factors associated with angiogenesis regulation and promotion. Further research is needed, especially in the mechanisms in which Pb-induced vascular endothelial growth factor (VEGF) disregulation is present. We believe that characterizing the connection between Pb and angiogenesis will provide helpful information for the development of intervention strategies to reduce the adverse effects of Pb exposure.


Asunto(s)
Neoplasias , Factor A de Crecimiento Endotelial Vascular , Humanos , Plomo/toxicidad , Neovascularización Patológica/inducido químicamente , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factores de Crecimiento Endotelial Vascular
6.
PLoS One ; 16(12): e0260793, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34855884

RESUMEN

Retinopathy of prematurity (ROP) is one of the main causes of blindness in children worldwide. Brain-derived neurotrophic factor (BDNF) and its receptor, tropomyosin-related kinase B (TrkB), play critical protective roles in the development and function of neurons and vasculature. Lack of BDNF expression results in increased endothelial cell apoptosis and reduced endothelial cell-cell contact. Premature babies who develop ROP tend to have lower serum BDNF levels. BDNF expression is also significantly lower in mouse retinas following exposure to hyperoxia compared to those reared in room air. Specifically, BDNF promotes angiogenic tube formation of endothelial cells (EC), and it is considered an EC survival factor required for stabilization of intramyocardial vessels. We hypothesized that the activation of TrkB receptor protects retinal vasculature in the mice during oxygen-induced ischemic retinopathy (OIR), a model of ROP. To test this hypothesis, we treated neonatal mice with 7,8-dihydroxyflavone (DHF) (5 mg/kg body weight), a TrkB receptor agonist. We examined its potential protective effects on retinal vessel obliteration and neovascularization, two hallmarks of ROP and OIR. We found that retinas from DHF treated postnatal day 8 (P8) and P12 mice have similar levels of vessel obliteration as retinas from age-matched control mice subjected to OIR. Similarly, DHF showed no significant effect on mitigation of retinal neovascularization during OIR in P17 mice. Collectively, our studies demonstrate that the TrkB receptor agonist DHF provides no significant protective effects during OIR.


Asunto(s)
Flavonas/farmacología , Isquemia/patología , Neovascularización Patológica/patología , Oxígeno/toxicidad , Receptor trkB/agonistas , Neovascularización Retiniana/patología , Retinopatía de la Prematuridad/patología , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Isquemia/inducido químicamente , Isquemia/tratamiento farmacológico , Isquemia/metabolismo , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neovascularización Retiniana/inducido químicamente , Neovascularización Retiniana/tratamiento farmacológico , Neovascularización Retiniana/metabolismo , Retinopatía de la Prematuridad/inducido químicamente , Retinopatía de la Prematuridad/tratamiento farmacológico , Retinopatía de la Prematuridad/metabolismo
7.
Int J Mol Sci ; 22(22)2021 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-34830344

RESUMEN

Electronic cigarettes are frequently viewed as a safer alternative to conventional cigarettes; however, evidence to support this perspective has not materialized. Indeed, the current literature reports that electronic cigarette use is associated with both acute lung injury and subclinical dysfunction to the lung and vasculature that may result in pathology following chronic use. E-cigarettes can alter vascular dynamics, polarize innate immune populations towards a proinflammatory state, compromise barrier function in the pulmonary endothelium and epithelium, and promote pre-oncogenic phenomena. This review will summarize the variety of e-cigarette products available to users, discuss current challenges in e-cigarette study design, outline the range of pathologies occurring in cases of e-cigarette associated acute lung injury, highlight disease supporting tissue- and cellular-level changes resulting from e-cigarette exposure, and briefly examine how these changes may promote tumorigenesis. Continued research of the mechanisms by which e-cigarettes induce pathology benefit users and clinicians by resulting in increased regulation of vaping devices, informing treatments for emerging diseases e-cigarettes produce, and increasing public awareness to reduce e-cigarette use and the onset of preventable disease.


Asunto(s)
Lesión Pulmonar Aguda/patología , Enfermedades Cardiovasculares/patología , Sistemas Electrónicos de Liberación de Nicotina , Neoplasias Pulmonares/patología , Vapeo/patología , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/inmunología , Animales , Plaquetas/efectos de los fármacos , Plaquetas/inmunología , Plaquetas/patología , Carcinogénesis/inmunología , Carcinogénesis/patología , Enfermedades Cardiovasculares/inducido químicamente , Enfermedades Cardiovasculares/inmunología , Citocinas/biosíntesis , Citocinas/inmunología , Humanos , Inmunidad Innata/efectos de los fármacos , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/patología , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/inmunología , Neovascularización Patológica/patología , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/patología , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/patología , Roedores , Vapeo/inmunología
8.
Bioengineered ; 12(1): 7508-7518, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34608841

RESUMEN

Hsa_circ_0001850_circ_0001850 (circ-UBAP2) is reported to be upregulated in diabetic retinopathy (DR). However, its role in high glucose (HG)-triggered oxidative stress and vascular dysfunction in DR is unclear. This study aimed to investigate the potential of circUBAP2 in DR. The content of malondialdehyde (MDA), and the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-PX) were analyzed using the corresponding kits. Western blotting was performed to detect the protein expression of Nrf2, HO-1, and SOD-1. MTT assay was conducted to assess cell viability. A transwell migration assay was used to determine the migration ability of human retinal microvascular endothelial cells (hRMECs). A Matrigel tube formation assay was performed to analyze tube formation. The targeting relationships were verified using a luciferase reporter assay. We found that the circ-UBAP2 expression increased in DR patients and HG-treated hRMECs. Downregulation of circ-UBAP2 ameliorated HG-induced oxidative stress and dysfunction of hRMECs. Mechanistically, circ-UBAP2 sponges miR-589-5p, which is downregulated under hyperglycemic conditions. In addition, EGR1 was confirmed to be a target gene of miR-589-5p and was overexpressed in HG-treated hRMECs. In addition, EGR1 reversed the effects of miR-589-5p and induced oxidative stress and dysfunction in hRMECs. Taken together, knockdown of circ-UBAP2 relieved HG-induced oxidative stress and dysfunctions of the hRMECs through the miR-589-5p/EGR1 axis, which may offer a promising therapeutic target for DR.


Asunto(s)
Retinopatía Diabética/metabolismo , MicroARNs/genética , Estrés Oxidativo/genética , ARN Circular/genética , Vasos Retinianos/citología , Células Cultivadas , Regulación hacia Abajo/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Endotelio Vascular/citología , Glucosa/efectos adversos , Humanos , MicroARNs/metabolismo , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/genética , Estrés Oxidativo/efectos de los fármacos , ARN Circular/metabolismo
9.
Exp Mol Pathol ; 123: 104715, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34699901

RESUMEN

This study was intended (1) to develop a robust animal model for hepatocellular carcinoma (HCC) research, in which HCC tumors develop in a background of fibrosis or cirrhosis; and (2) to explore time-dependent regulatory changes in key molecular markers during disease advancement and HCC development. With the aim of establishing such HCC model, male Sprague-Dawley rats were injected with diethylnitrosamine (DEN) at a dose of 30 mg/kg twice a week for 10 weeks then once a week from 12th to 16th weeks. The rats were kept under observation until 18th week. At defined time intervals (2nd, 4th, 12th, and 18th week), serum biomarkers and microscopic components of tissue samples were used to investigate the chronic progression of liver disease, while gene and protein analysis was used to monitor expression patterns during HCC development. DEN-intoxicated rats manifested inflammation at week 4, fibrosis at week 12 and cirrhosis with early HCC tumors at week 18. Molecular analysis revealed that key markers of inflammation (Il-1ß, Il-6, and Tnf-α), fibrosis (Tgf-ß1, Col1α1, Col3α1, and Timp-1), and angiogenesis (Hif1-α and Vegf) were promptly (P ≤ 0.001) up-regulated at week 4, week 12 and week 18, respectively. Oxidative stress (iNos, Cyp2e1, and Sod1) and pro-apoptotic (Bax) markers showed significant upregulation from week 4 to week 12. However, Sod1 and Bax expressions dropped after week 12 and reached a minimum at 18th week. Strikingly, expressions of anti-apoptotic (Bcl-2) and cell proliferation (Pcna, Hgf, and Afp) markers were abruptly increased at week 18. Collectively, we describe an 18-week HCC model in DEN-intoxicated rats that exhibit chronic inflammation, oxidative imbalance, advance fibrosis/cirrhosis, halted apoptosis, and angiogenic sprouting, progressively.


Asunto(s)
Carcinogénesis/genética , Carcinoma Hepatocelular/genética , Inflamación/genética , Neoplasias Hepáticas/genética , Proteínas de Neoplasias/genética , Animales , Apoptosis/genética , Carcinoma Hepatocelular/inducido químicamente , Carcinoma Hepatocelular/patología , Proliferación Celular/genética , Dietilnitrosamina/toxicidad , Modelos Animales de Enfermedad , Fibrosis/inducido químicamente , Fibrosis/genética , Fibrosis/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Inflamación/inducido químicamente , Inflamación/patología , Hígado/efectos de los fármacos , Hígado/patología , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/patología , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Ratas
10.
Oxid Med Cell Longev ; 2021: 5520644, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34457115

RESUMEN

Age-related macular degeneration (AMD) is a common and severe blinding disease among people worldwide. Retinal inflammation and neovascularization are two fundamental pathological processes in AMD. Recent studies showed that P2X7 receptor was closely involved in the inflammatory response. Here, we aim to investigate whether A740003, a P2X7 receptor antagonist, could prevent retinal inflammation and neovascularization induced by oxidized low-density lipoprotein (ox-LDL) and explore the underlying mechanisms. ARPE-19 cells and C57BL/6 mice were treated with ox-LDL and A740003 successively for in vitro and in vivo studies. In this research, we found that A740003 suppressed reactive oxygen species (ROS) generation and inhibited the activation of Nod-like receptor pyrin-domain protein 3 (NLRP3) inflammasome and nuclear factor-κB (NF-κB) pathway. A740003 also inhibited the generation of angiogenic factors in ARPE-19 cells and angiogenesis in mice. The inflammatory cytokines and phosphorylation of inhibitor of nuclear factor-κB alpha (IKBα) were repressed by A740003. Besides, ERG assessment showed that retinal functions were remarkably preserved in A740003-treated mice. In summary, our results revealed that the P2X7 receptor antagonist reduced retinal inflammation and neovascularization and protected retinal function. The protective effects were associated with regulation of NLRP3 inflammasome and the NF-κB pathway, as well as inhibition of angiogenic factors.


Asunto(s)
Inflamación/tratamiento farmacológico , Lipoproteínas LDL/toxicidad , Neovascularización Patológica/tratamiento farmacológico , Estrés Oxidativo , Antagonistas del Receptor Purinérgico P2X/farmacología , Receptores Purinérgicos P2X7/química , Retinitis/tratamiento farmacológico , Animales , Citocinas/metabolismo , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/genética , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Especies Reactivas de Oxígeno/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Retinitis/inducido químicamente , Retinitis/metabolismo , Retinitis/patología , Transducción de Señal
11.
Semin Cancer Biol ; 76: 279-286, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34428550

RESUMEN

MicroRNAs (miRNAs) are small endogenous non-coding RNAs that regulate cancer initiation, development, angiogenesis, and therapeutic resistance. Metal exposure widely occurs through air, water, soil, food, and industrial contaminants. Hundreds of millions of people may have metal exposure associated with toxicity, serious health problems, and cancer occurrence. Metal exposure is found to induce oxidative stress, DNA damage and repair, and activation of multiple signaling pathways. However, molecular mechanisms of metal-induced carcinogenesis remain to be elucidated. Recent studies demonstrated that the exposure of metals such as arsenic, hexavalent chromium, cadmium, and nickel caused dysregulation of microRNAs that are implicated to play an important role in cell transformation, tumor growth and angiogenesis. This review focuses on the recent studies that show metal-induced miRNA dysregulation and underlined mechanisms in cell malignant transformation, angiogenesis and tumor growth.


Asunto(s)
Transformación Celular Neoplásica/inducido químicamente , Metales/efectos adversos , MicroARNs , Neovascularización Patológica/inducido químicamente , Animales , Humanos
12.
Int J Mol Sci ; 22(14)2021 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-34299320

RESUMEN

Chemotherapy is still widely used as a coadjutant in gastric cancer when surgery is not possible or in presence of metastasis. During tumor evolution, gatekeeper mutations provide a selective growth advantage to a subpopulation of cancer cells that become resistant to chemotherapy. When this phenomenon happens, patients experience tumor recurrence and treatment failure. Even if many chemoresistance mechanisms are known, such as expression of ATP-binding cassette (ABC) transporters, aldehyde dehydrogenase (ALDH1) activity and activation of peculiar intracellular signaling pathways, a common and universal marker for chemoresistant cancer cells has not been identified yet. In this study we subjected the gastric cancer cell line AGS to chronic exposure of 5-fluorouracil, cisplatin or paclitaxel, thus selecting cell subpopulations showing resistance to the different drugs. Such cells showed biological changes; among them, we observed that the acquired chemoresistance to 5-fluorouracil induced an endothelial-like phenotype and increased the capacity to form vessel-like structures. We identified the upregulation of thymidine phosphorylase (TYMP), which is one of the most commonly reported mutated genes leading to 5-fluorouracil resistance, as the cause of such enhanced vasculogenic ability.


Asunto(s)
Resistencia a Antineoplásicos , Fluorouracilo/farmacología , Neovascularización Patológica/inducido químicamente , Neoplasias Gástricas/irrigación sanguínea , Neoplasias Gástricas/tratamiento farmacológico , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Cisplatino/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Fluorouracilo/metabolismo , Humanos , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Paclitaxel/farmacología , Neoplasias Gástricas/patología , Talidomida/farmacología , Timidina Fosforilasa/genética , Regulación hacia Arriba/efectos de los fármacos
13.
Front Immunol ; 12: 649591, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33995368

RESUMEN

Psoriasis is a chronic proliferative autoimmune dermatologic disease characterised by abnormal angiogenesis. Thus, regulating angiogenesis in the skin is an important treatment strategy for psoriasis. PSORI-CM02, an empirical Chinese medicine formula optimised from Yin Xie Ling, was created by the Chinese medicine specialist, Guo-Wei Xuan. Clinical studies have shown that PSORI-CM02 is safe and effective for the treatment of psoriasis. However, its anti-psoriatic mechanisms remain to be further explored. In this study, we investigated the effects of PSORI-CM02 on angiogenesis in the skin and the underlying mechanisms in IL-17A-stimulated human umbilical vein endothelial cells (HUVECs) and a murine model of imiquimod (IMQ)-induced psoriasis. In vitro, PSORI-CM02 significantly inhibited the proliferation and migration of IL-17A-stimulated HUVECs in a dose-dependent manner. Further, it markedly regulated the antioxidative/oxidative status and inflammation; suppressed the expression of VEGF, VEGFR1, VEGFR2, ANG1, and HIF-1α; and reduced the phosphorylation of MAPK signalling pathway components in IL-17A-stimulated HUVECs. In vivo studies showed that PSORI-CM02 markedly reduced angiogenesis in the skin of mice with IMQ-induced psoriasis, while significantly rebalancing antioxidant/oxidant levels; inhibiting the production of IL-6, TNF-α, IL-17A, and IL-17F; and repressing the synthesis of angiogenic mediators. In addition, PSORI-CM02 markedly reduced the activation of the MAPK signalling pathway in psoriatic skin tissue. Taken together, our results demonstrated that PSORI-CM02 inhibited psoriatic angiogenesis by reducing the oxidative status and inflammation, suppressing the expression of angiogenesis-related molecules, and inhibiting the activation of the MAPK signalling pathway in vitro and in vivo.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Neovascularización Patológica/tratamiento farmacológico , Psoriasis/tratamiento farmacológico , Piel/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/uso terapéutico , Células Endoteliales de la Vena Umbilical Humana , Humanos , Imiquimod/administración & dosificación , Imiquimod/toxicidad , Interleucina-17/inmunología , Interleucina-17/metabolismo , Queratinocitos , Masculino , Ratones , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/inmunología , Neovascularización Patológica/patología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/inmunología , Psoriasis/inducido químicamente , Psoriasis/inmunología , Psoriasis/patología , Piel/irrigación sanguínea , Piel/inmunología , Piel/patología
14.
Int Immunopharmacol ; 96: 107660, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33862553

RESUMEN

Alpinetin is the major active ingredient of Alpiniakatsumadai Hayata. As a kind of novel plant-derived flavonoid, alpinetin has shown potent hepatoprotective effect against many liver diseases such as non-alcoholic fatty liver and lipopolysaccharide/d-Galactosamine-induced liver injury. However, its roles in liver fibrosis remain to be determined. The aim of the current study was to investigate the effect of alpinetin in mice with carbon tetrachloride (CCl4)-induced liver fibrosis, and to elucidate the underlying mechanisms of action. Alpinetin ameliorated the CCl4-induced liver injury and fibrosis in mice, as shown by decreased collagen deposition and the decreased expression of liver fibrosis marker proteins. Alpinetin suppressed the inflammation and oxidative stress in fibrotic livers of mice, as evidenced by decreased levels of proinflammatory factors, the decreased reactive oxygen species (ROS) and malondialdehyde (MDA) levels, and the increased activities of antioxidant enzymes. In addition, alpinetin attenuated the angiogenesis in fibrotic livers of the test animals. Mechanistically, alpinetin inhibited the CCl4-induced expression of NLRP3, ASC, cleaved caspase-1, mature (cleaved-) IL-1ß, and IL-18 in livers of mice. Furthermore, alpinetin resulted in an increased in the nuclear expression and a decrease in the cytoplasmic expression of Nrf2, as well as increased protein expression of downstream target enzymes, GCLC, HO-1, NQO1, and GCLM, thus exerting the antioxidant effect. Overall, these findings suggested that the anti-fibrotic effect of alpinetin can be attributed to the inhibition of NLRP3-mediated anti-inflammatory activities and Nrf2-mediated anti-oxidative activities, in addition to the decrement of hepatic angiogenesis.


Asunto(s)
Antiinflamatorios/farmacología , Antioxidantes/farmacología , Flavanonas/farmacología , Cirrosis Hepática/tratamiento farmacológico , Factor 2 Relacionado con NF-E2/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Neovascularización Patológica/tratamiento farmacológico , Animales , Antiinflamatorios/uso terapéutico , Antioxidantes/uso terapéutico , Tetracloruro de Carbono/toxicidad , Colágeno/efectos de los fármacos , Colágeno/metabolismo , Modelos Animales de Enfermedad , Flavanonas/uso terapéutico , Inflamasomas/efectos de los fármacos , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/patología , Masculino , Ratones Endogámicos C57BL , Neovascularización Patológica/inducido químicamente , Estrés Oxidativo/efectos de los fármacos , Oxidorreductasas/efectos de los fármacos , Oxidorreductasas/metabolismo , Transducción de Señal/efectos de los fármacos
15.
Sci Rep ; 11(1): 6216, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33737643

RESUMEN

Vascular Endothelial Growth Factor (VEGF), a key mediator of angiogenesis and vascular repair, is reduced in chronic ischemic renal diseases, leading to microvascular rarefaction and deterioration of renal function. We developed a chimeric fusion of human VEGF-A121 with the carrier protein Elastin-like Polypeptide (ELP-VEGF) to induce therapeutic angiogenesis via targeted renal VEGF therapy. We previously showed that ELP-VEGF improves renal vascular density, renal fibrosis, and renal function in swine models of chronic renal diseases. However, VEGF is a potent cytokine that induces angiogenesis and increases vascular permeability, which could cause undesired off-target effects or be deleterious in a patient with a solid tumor. Therefore, the current study aims to define the toxicological profile of ELP-VEGF and assess its risk for exacerbating tumor progression and vascularity using rodent models. A dose escalating toxicology assessment of ELP-VEGF was performed by administering a bolus intravenous injection at doses ranging from 0.1 to 200 mg/kg in Sprague Dawley (SD) rats. Blood pressure, body weight, and glomerular filtration rate (GFR) were quantified longitudinally, and terminal blood sampling and renal vascular density measurements were made 14 days after treatment. Additionally, the effects of a single administration of ELP-VEGF (0.1-10 mg/kg) on tumor growth rate, mass, and vascular density were examined in a mouse model of breast cancer. At doses up to 200 mg/kg, ELP-VEGF had no effect on body weight, caused no changes in plasma or urinary markers of renal injury, and did not induce renal fibrosis or other histopathological findings in SD rats. At the highest doses (100-200 mg/kg), ELP-VEGF caused an acute, transient hypotension (30 min), increased GFR, and reduced renal microvascular density 14 days after injection. In a mouse tumor model, ELP-VEGF did not affect tumor growth rate or tumor mass, but analysis of tumor vascular density by micro-computed tomography (µCT) revealed significant, dose dependent increases in tumor vascularity after ELP-VEGF administration. ELP-VEGF did not induce toxicity in the therapeutic dosing range, and doses one hundred times higher than the expected maximum therapeutic dose were needed to observe any adverse signs in rats. In breast tumor-bearing mice, ELP-VEGF therapy induced a dose-dependent increase in tumor vascularity, demanding caution for potential use in a patient suffering from kidney disease but with known or suspected malignancy.


Asunto(s)
Productos Biológicos/farmacología , Neoplasias de la Mama/irrigación sanguínea , Elastina/genética , Neovascularización Patológica/inducido químicamente , Proteínas Recombinantes de Fusión/farmacología , Insuficiencia Renal Crónica/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Productos Biológicos/metabolismo , Presión Sanguínea/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/patología , Permeabilidad Capilar/efectos de los fármacos , Modelos Animales de Enfermedad , Elastina/metabolismo , Femenino , Expresión Génica , Tasa de Filtración Glomerular/efectos de los fármacos , Xenoinjertos , Humanos , Hipotensión/inducido químicamente , Hipotensión/diagnóstico por imagen , Hipotensión/fisiopatología , Ratones , Imitación Molecular , Neovascularización Patológica/diagnóstico por imagen , Neovascularización Patológica/patología , Neovascularización Fisiológica/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Insuficiencia Renal Crónica/diagnóstico por imagen , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/fisiopatología , Porcinos , Pruebas de Toxicidad Crónica , Factor A de Crecimiento Endotelial Vascular/metabolismo , Microtomografía por Rayos X
16.
Oncol Rep ; 45(5)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33760153

RESUMEN

Mineral dust­induced gene (mdig) is a novel lung cancer­related oncogene. The aim of this study was to explore the effects of mdig on angiogenesis and lymphangiogenesis by vascular endothelial growth factor (VEGF) in lung adenocarcinoma. mdig­overexpressing A549, H1299 and 293T cells, mdig­silenced A549, human umbilical vein endothelial cells (HUVECs) and human lymphatic endothelial cells (HLECs) were cultured under normoxic and hypoxic conditions. Protein expression levels of mdig, epidermal growth factor receptor (EGFR), phospho(p)­EGFR Tyr1068, hypoxia­inducible factor­1α (HIF­1α), VEGF­A/C/D and VEGF­R1/R2/R3 were assessed using western blotting. mRNA expression levels of mdig, EGFR and HIF­1α were measured using RT­qPCR. Tube formation and xenograft tumor experiments were performed to examine the mechanism of mdig in angiogenesis and lymphangiogenesis. Protein expression levels of EGFR, HIF­1α and VEGF­A/C/D were significantly upregulated in cells cultured under hypoxic conditions compared with those cultured under normoxic conditions, whereas the levels of mdig were decreased. Protein expression levels of EGFR, p­EGFR and VEGF­A/R1/R2 were significantly increased in the mdig­overexpressing cells, whereas the levels of HIF­1α and VEGF­C/D/R3 were decreased compared with those in control cells, all of which were reversed in mdig­silenced cells. Tumor volumes and density of angiogenesis in the mdig­overexpressing group were significantly increased compared with those in the control group, whereas the density of lymphangiogenesis was decreased. No tumors formed in the mdig­silenced group after 3 weeks of assessment in vivo. Protein expression levels of EGFR, p­EGFR, VEGF­A and angiogenesis density were significantly reduced in the mdig­overexpressing cells treated with an EGFR inhibitor, whereas the levels of HIF­1α, VEGF­C/D and the lymphangiogenesis density were significantly increased in mdig­overexpressing cells treated with a HIF­1α agonist. All changes in protein expression were reversed in EGFR agonist and HIF­1α inhibitor treated mdig­silenced cells. In conclusion, mdig is an oxygen­sensitive protein that promotes tumor growth and angiogenesis by activating the EGFR/p­EGFR/VEGF­A/VEGF­R1/R2 pathway and inhibits lymphangiogenesis by blocking the HIF­1α/VEGF­C/D/VEGF­R3 pathway.


Asunto(s)
Adenocarcinoma del Pulmón/patología , Dioxigenasas/metabolismo , Histona Demetilasas/metabolismo , Neoplasias Pulmonares/patología , Linfangiogénesis/genética , Neovascularización Patológica/genética , Proteínas Nucleares/metabolismo , Adenocarcinoma del Pulmón/irrigación sanguínea , Adenocarcinoma del Pulmón/genética , Animales , Línea Celular Tumoral , Desmetilación del ADN , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Histonas/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/agonistas , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Pulmón/irrigación sanguínea , Pulmón/patología , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/genética , Linfangiogénesis/efectos de los fármacos , Ratones , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/patología , Transducción de Señal/genética , Carga Tumoral , Hipoxia Tumoral/genética , Regulación hacia Arriba , Factores de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Am J Chin Med ; 49(3): 737-751, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33683188

RESUMEN

Angiogenesis plays a crucial role in tumor growth and metastasis. Vascular endothelial growth factor (VEGF)-stimulated endothelial cell proliferation and migration are critical steps in tumor angiogenesis. Here, we investigated the anti-angiogenic activity of xanthorrhizol, a sesquiterpenoid isolated from the Indonesian medicinal plant Curcuma xanthorrhiza. Xanthorrhizol at noncytotoxic concentrations inhibited the proliferation, migration, and formation of capillary-like tubes in VEGF-treated human umbilical vein endothelial cells (HUVECs). Xanthorrhizol inhibited the phosphorylation of Akt and endothelial nitric oxide synthase (eNOS) and the expression of vascular cell adhesion molecule (VCAM)-1 and E-selectin in VEGF-treated HUVECs. The expression and transcriptional activity of NF-[Formula: see text]B were downregulated by xanthorrhizol in VEGF-treated HUVECs. Furthermore, xanthorrhizol significantly inhibited VEGF-induced angiogenesis in the chorioallantoic membrane of fertilized eggs and Matrigel plugs subcutaneously injected into mice. Xanthorrhizol inhibited tumor volume and tumor-derived angiogenesis in mice inoculated with breast cancer cells. The in vitro and in vivo anti-angiogenic activities of xanthorrhizol were as potent as those of curcumin, a well-known anticancer agent derived from C. longa. Taken together, xanthorrhizol inhibits VEGF-induced angiogenesis of endothelial cells by blocking the activation of the PI3K/Akt/eNOS axis and subsequent upregulation of adhesion molecules induced by the transcriptional activation of NF-[Formula: see text]B. Xanthorrhizol is a promising anti-angiogenic agent and can serve as a beneficial agent to enhance anticancer treatments.


Asunto(s)
Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , FN-kappa B/metabolismo , Neovascularización Patológica/genética , Neovascularización Patológica/prevención & control , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fenoles/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factor A de Crecimiento Endotelial Vascular/efectos adversos , Animales , Curcuma/química , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Neovascularización Patológica/inducido químicamente , Fenoles/aislamiento & purificación , Fenoles/uso terapéutico , Fosfatidilinositol 3-Quinasas/metabolismo , Fitoterapia , Células Tumorales Cultivadas
18.
Angiogenesis ; 24(3): 567-581, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33550461

RESUMEN

OBJECTIVE: Statins pleiotropically provide additional benefits in reducing atherosclerosis, but their effects on intraplaque angiogenesis (IPA) and hemorrhage (IPH) remain unclear. Therefore, we discriminated statin's lipid-lowering dependent and independent effects on IPA and IPH. APPROACH AND RESULTS: ApoE3*Leiden mice are statin-responsive due to ApoE and LDLR presence, but also allow to titrate plasma cholesterol levels by diet. Therefore, ApoE3*Leiden mice were fed a high-cholesterol-inducing-diet (HCD) with or without atorvastatin (A) or a moderate-cholesterol-inducing-diet (MCD). Mice underwent vein graft surgery to induce lesions with IPA and IPH. Cholesterol levels were significantly reduced in MCD (56%) and HCD + A (39%) compared to HCD with no significant differences between MCD and HCD + A. Both MCD and HCD + A have a similar reduction in vessel remodeling and inflammation comparing to HCD. IPA was significantly decreased by 30% in HCD + A compared to HCD or MCD. Atorvastatin treatment reduced the presence of immature vessels by 34% vs. HCD and by 25% vs. MCD, resulting in a significant reduction of IPH. Atorvastatin's anti-angiogenic capacity was further illustrated by a dose-dependent reduction of ECs proliferation and migration. Cultured mouse aortic-segments lost sprouting capacity upon atorvastatin treatment and became 30% richer in VE-Cadherin expression and pericyte coverage. Moreover, Atorvastatin inhibited ANGPT2 release and decreased VE-Cadherin(Y685)-phosphorylation in ECs. CONCLUSIONS: Atorvastatin has beneficial effects on vessel remodeling due to its lipid-lowering capacity. Atorvastatin has strong pleiotropic effects on IPA by decreasing the number of neovessels and on IPH by increasing vessel maturation. Atorvastatin improves vessel maturation by inhibiting ANGPT2 release and phospho(Y658)-mediated VE-Cadherin internalization.


Asunto(s)
Angiopoyetina 2 , Antígenos CD , Atorvastatina/farmacología , Cadherinas , Colesterol en la Dieta/efectos adversos , Neovascularización Patológica , Placa Aterosclerótica , Angiopoyetina 2/genética , Angiopoyetina 2/metabolismo , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Colesterol en la Dieta/farmacología , Masculino , Ratones , Ratones Mutantes , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Placa Aterosclerótica/inducido químicamente , Placa Aterosclerótica/tratamiento farmacológico , Placa Aterosclerótica/genética , Placa Aterosclerótica/metabolismo
19.
Toxicology ; 452: 152722, 2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33592256

RESUMEN

Aroclor 1254 is a mixture of polychlorinated biphenyls that are reported to disrupt thyroid hormone homeostasis, yet little is known on its effect on thyroid gland microarchitecture. Lycopene is a commonly used potent antioxidant. This study is a biochemical, histological, and immunohistochemical assessment of the effect of Aroclor 1254 on the morphology, proliferation, and angiogenesis of the thyroid gland in rat and to evaluate the possible ameliorating role of lycopene. Twenty-four adult male albino rats were divided into 4 groups; Control, lycopene-treated (4 mg/kg/day orally for 30 days), Aroclor 1254-treated (2 mg/kg/day intraperitoneally for 30 days), and lycopene & Aroclor 1254-treated group. Serum thyroid hormones, thyroid-stimulating hormone (TSH), and tissue malondialdehyde (MDA) were quantified. Thyroid specimens were processed for histological staining with hematoxylin and eosin, periodic acid-Schiff, and Mallory's trichrome stains as well as immunohistochemical staining for detection of calcitonin, Ki67, and VEGF. In this study, Aroclor 1254-treated animals recorded a significant decline in both serum T3 and T4 coupled with a significant elevation in both TSH and tissue MDA. Histological sections showed small irregular follicles with the formation of hyperplastic and micro follicles. Some follicular and parafollicular cells depicted nuclear and cytoplasmic alterations associating with scanty or absent colloid in addition to signs of inflammation and fibrosis. A significant upregulation in the immunohistochemical expression of calcitonin, Ki67, and VEGF was recorded. Lycopene co-treatment successfully reinstated the values of most studied parameters and retrieved a near-control thyroid morphology. In conclusion, Aroclor 1254 impacted the thyroid hormone homeostasis, morphology, proliferation, and angiogenesis of the thyroid gland in rat, while lycopene efficiently ameliorated these adverse effects.


Asunto(s)
Antioxidantes/uso terapéutico , Proliferación Celular/efectos de los fármacos , Licopeno/uso terapéutico , Neovascularización Patológica/tratamiento farmacológico , Glándula Tiroides/efectos de los fármacos , Animales , Antioxidantes/farmacología , Proliferación Celular/fisiología , Licopeno/farmacología , Masculino , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Ratas , Ratas Wistar , Glándula Tiroides/metabolismo , Glándula Tiroides/patología , Hormonas Tiroideas/metabolismo
20.
J Cell Mol Med ; 25(8): 4001-4013, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33609067

RESUMEN

Molecular targeted agents are pharmacologically used to treat liver fibrosis and have gained increased attention. The present study examined the preventive effect of lenvatinib on experimental liver fibrosis and sinusoidal capillarization as well as the in vitro phenotypes of hepatic stellate cells. LX-2, a human stellate cell line, was used for in vitro studies. In vivo liver fibrosis was induced in F344 rats using carbon tetrachloride by intraperitoneal injection for 8 weeks, and oral administration of lenvatinib was started two weeks after initial injection of carbon tetrachloride. Lenvatinib restrained proliferation and promoted apoptosis of LX-2 with suppressed phosphorylation of extracellular signal-regulated kinase 1/2 and AKT. It also down-regulated COL1A1, ACTA2 and TGFB1 expressions by inhibiting the transforming growth factor-ß1/Smad2/3 pathway. Treatment with lenvatinib also suppressed platelet-derived growth factor-BB-stimulated proliferation, chemotaxis and vascular endothelial growth factor-A production, as well as basic fibroblast growth factor-induced LX-2 proliferation. In vivo study showed that lenvatinib attenuated liver fibrosis development with reduction in activated hepatic stellate cells and mRNA expression of profibrogenic markers. Intrahepatic neovascularization was ameliorated with reduced hepatic expressions of Vegf1, Vegf2 and Vegfa in lenvatinib-treated rats. Collectively, these results suggest the potential use of lenvatinib as a novel therapeutic strategy for liver fibrosis.


Asunto(s)
Capilares/efectos de los fármacos , Células Estrelladas Hepáticas/efectos de los fármacos , Cirrosis Hepática/prevención & control , Hígado/efectos de los fármacos , Neovascularización Patológica/prevención & control , Compuestos de Fenilurea/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Quinolinas/farmacología , Animales , Capilares/metabolismo , Capilares/patología , Tetracloruro de Carbono/toxicidad , Células Cultivadas , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Hígado/irrigación sanguínea , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/patología , Masculino , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/patología , Ratas , Ratas Endogámicas F344
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...