Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(5)2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38473712

RESUMO

Canine-mammary-gland tumors (CMTs) are prevalent in female dogs, with approximately 50% of them being malignant and often presenting as inoperable owing to their size or metastasis. Owing to poor outcomes, effective alternatives to conventional chemotherapy for humans are necessary. Two estrogen receptors, estrogen receptor alpha (ERα) and estrogen receptor beta (ERß), which act in opposition to each other, are involved, and CMT growth involves ERα through the phosphoinositide 3-kinases (PI3K)/AKT pathway. In this study, we aimed to identify the synergistic anti-cancer effects of ERB-041, an ERß agonist, and genistein, an isoflavonoid from soybeans known to have ERß-specific pseudo-estrogenic actions, on CMT-U27 and CF41.Mg CMT cell lines. ERB-041 and genistein synergistically inhibited cell proliferation and increased the number of annexin V-positive cells in both cell lines. Furthermore, we observed a synergistic increase in the Bax/Bcl-2 ratio and cleaved caspase-3 expression. Additionally, cell-cycle arrest occurred through the synergistic regulation of cyclin D1 and cyclin-dependent kinase 4 (CDK4). We also found a synergistic decrease in the expression of ERα, and the expression of proteins involved in the PI3K/AKT pathway, including p-PI3K, phosphatase and tensin homolog (PTEN), AKT, and mechanistic target of rapamycin (mTOR). In conclusion, ERB-041 and genistein exhibited a synergistic anticancer effect on CMTs, suggesting that cotreatment with ERB-041 and genistein is a promising treatment for CMTs.


Assuntos
Glândulas Mamárias Humanas , Oxazóis , Receptores de Estrogênio , Cães , Animais , Feminino , Humanos , Receptores de Estrogênio/metabolismo , Genisteína/farmacologia , Receptor beta de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Regulação para Baixo , Glândulas Mamárias Humanas/metabolismo , Estrogênios/metabolismo
2.
BMC Vet Res ; 19(1): 223, 2023 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-37880653

RESUMO

Canine mammary gland tumors (CMTs) are the most common and lethal cancers in female dogs. Dysregulated phosphoinositide 3-kinases (PI3K)/AKT pathway reportedly was involved in the growth and metastasis of CMTs. However, there are few studies on therapeutic strategies for targeting the PI3K pathway in CMTs. In this study, we aimed to determine whether palmatine, a natural isoquinoline alkaloid with anti-cancer properties, could inhibit the growth of CMTs and whether the inhibitory effect was mediated through the PI3K/AKT pathway. Our in vitro experiments on CMT-U27, a CMT cell line, showed that palmatine reduced cell proliferation and induced cell death. Western blotting results revealed that palmatine decreased the protein expression of PI3K, PTEN, AKT, and mechanistic target of rapamycin in the PI3K/AKT pathway, which was supported by the results of immunocytochemistry. Additionally, palmatine suppressed the migration and tube formation of canine aortic endothelial cells as well as the migration of CMT U27 cells. Our in vivo results showed that palmatine inhibited tumor growth in a CMT-U27 mouse xenograft model. We observed a decreased expression of proteins in the PI3K/AKT pathway in tumor tissues, similar to the in vitro results. Furthermore, palmatine significantly disrupted the tumor vasculature and inhibited metastasis to adjacent lymph nodes. In conclusion, our findings demonstrate that palmatine exerts anti-cancer effects against CMTs by inhibiting PI3K/AKT signaling pathway, suggesting that palmatine has potential as a canine-specific PI3K inhibitor for the treatment of CMTs.


Assuntos
Glândulas Mamárias Humanas , Fosfatidilinositol 3-Quinases , Cães , Animais , Feminino , Camundongos , Humanos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-akt/farmacologia , Células Endoteliais/metabolismo , Glândulas Mamárias Humanas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células
3.
Int J Vet Sci Med ; 11(1): 87-93, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37649734

RESUMO

Canine C-reactive protein (cCRP) is one of the major positive acute phase proteins in dogs and is commonly measured to detect and monitor systemic inflammation as well as the efficacy of treatment. Traditional methods for testing cCPR, including enzyme-linked immunosorbent assay (ELISA), have some drawbacks, such as a long time for diagnosis and the requirement of well-equipped laboratories. Therefore, there is a need for a rapid and precise diagnostic test for cCRP at point-of-care. This study assessed the accuracy, precision, and validated clinical effectiveness of a diagnostic test based on fluorescent lateral flow immunoassay to detect cCRP. For the standard cCRP concentration ranging from 0 to 200 µg/mL, the cCRP diagnostic test showed strong linearity with R2 of 0.9977 (p < 0.001), and both inter- and intra-assay CVs were <14%. The limit of detection and limit of quantitation were found to be 4.0 µg/mL and 5.0 µg/mL, respectively. The cCRP serum concentration was evaluated in 21 client-owned dogs and the results were compared to a previously validated ELISA. The Pearson Correlation Coefficient between the diagnostic test kit and ELISA was 0.942 [95% confidence interval: 0.859 to 0.976, p < 0.001], and the Bland-Altman plot indicated a bias of 26.82% [95% limits of agreement: -56.03 to 109.67], indicating a significant correlation and the agreement between the data from the cCRP diagnostic test and ELISA. In conclusion, the fluorescent immunoassay based diagnostic test is a suitable option for rapidly and precisely detecting cCRP in dogs, providing a convenient alternative to traditional methods for diagnosing acute inflammation.

4.
Vet Sci ; 10(2)2023 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-36851388

RESUMO

Mammary gland tumors are the most common neoplasms in female dogs, of which 50% are malignant. Esculetin, a coumarin derivative, reportedly induces death in different types of cancer cells. In this study, we explore the anticancer effects of esculetin against CMT-U27 and CF41.mg canine mammary gland tumor cells. Esculetin significantly inhibited the viability and migration of both CMT-U27 and CF41.mg cells in a dose- and time-dependent manner. Flow cytometric analysis and terminal deoxynucleotidyl transferase dUTP nick-end labeling assay revealed increased numbers of annexin-V-positive cells and DNA fragmentation. Furthermore, a cell cycle analysis demonstrated that esculetin blocked the cell progression at the G0/G1 phase and the S phase in CMT-U27 and CF41.mg cells. These results were supported by a Western blot analysis, which revealed upregulated protein expression of cleaved caspase-3, a marker of apoptosis, and downregulated cyclin-dependent kinase 4 and cyclin D1 protein, the cell cycle regulators. In conclusion, this novel study proves that esculetin exerts in vitro antitumor effects by inducing apoptosis and cell cycle arrest in canine mammary gland tumors.

5.
Artigo em Inglês | MEDLINE | ID: mdl-36110191

RESUMO

Methyl gallate is a phenolic compound mainly found in medicinal plants. It has been reported to its anticancer activity in various tumors. In this study, we aimed to demonstrate the antitumor effect of methyl gallate in the melanoma mouse model and B16F10 cells. Our results showed that methyl gallate decreased cell viability and induced apoptosis by increasing the expression of cleaved caspase3 in B16F10 cells and prevented cell migration and tube formation in human umbilical vein endothelial cells. In B16F10 cell-inoculated mice, methyl gallate not only decreased tumor volume by 30% but also significantly reduced tumor vessel density and pericyte coverage. Moreover, methyl gallate diminished by close to 50% the expression of cytokeratin and LYVE-1 in mouse right inguinal lymph nodes, indicating that methyl gallate could suppress metastasis. In conclusion, this study suggests that methyl gallate inhibits tumor development by inducing apoptosis and blocking tumor angiogenesis and metastasis and might be considered a therapeutic agent for melanoma.

6.
Animals (Basel) ; 11(6)2021 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-34071074

RESUMO

Fucoxanthin is a carotenoid derived from brown algae. It is known to exhibit anticancer activity, including the promotion of apoptosis and cell cycle arrest in several tumors. However, it remains unclear whether fucoxanthin exhibits anticancer activity against mammary gland tumors. In this study, we evaluated fucoxanthin activity against canine mammary tumor cells (CMT-U27) and human umbilical vein endothelial cells (HUVECs) to investigate its effect on cell viability, migration, tube formation, and angiopoietin 2 (Ang2) expression. Our results showed that fucoxanthin induced apoptosis via caspase activation in CMT-U27 cells. In rat aortic ring assay, fucoxanthin suppressed endothelial cell sprouting. Furthermore, fucoxanthin inhibited tube formation and migration in HUVECs. The number of migrated cells was assessed using CMT-U27 cells. The results demonstrated that fucoxanthin exerted anti-angiogenic activity on HUVECs and CMT-U27 cells by promoting Ang2 expression. In conclusion, our results demonstrated that fucoxanthin induced tumor cell death and inhibited angiogenesis, suggesting that fucoxanthin could be considered as a promising therapeutic agent for canine mammary gland tumors.

7.
Int J Med Sci ; 17(18): 3049-3057, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33173425

RESUMO

Malignant melanoma is one of the most deadly skin cancer, due to its aggressive proliferation and metastasis. Naringenin, abundantly present in citrus fruits, has widely studied in cancer therapy. In this study, we investigated whether naringenin also has anticancer effects against B16F10 murine and SK-MEL-28 human melanoma cells. Moreover, we assessed the effects of naringenin treatment on angiogenesis of HUVECs and ex vivo sprouting of microvessels.Naringenin inhibited tumor cell proliferation and migration in a dose-dependent manner in B16F10 and SK-MEL-28 cells, which is supported by the results that phosphorylation of ERK1/2 and JNK MAPK decreased. Furthermore, naringenin induced cell apoptosis. Western blot analysisshowed naringenin treatment significantly upregulated the protein expression of activated cas3 and PARP in B16F10 and SK-MEL-28 cells. In addition, in vitro and ex vivo angiogenesis assays demonstrated that naringenin treatment potently suppressed EC migration, tube formation, and sprouting of microvessels. RT-PCR analysis showed that naringenin treatment significantly reduced the mRNA expression of Tie2, but did not inhibit the expression of Ang2. In conclusion, present study demonstrates the anticancer effects of naringenin by its induction of tumor cell death and inhibition of angiogenesis in malignant melanoma, suggesting that naringenin has potential as a safe and effective therapeutic agent to treat melanoma.


Assuntos
Inibidores da Angiogênese/farmacologia , Flavanonas/farmacologia , Melanoma Experimental/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , Inibidores da Angiogênese/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Flavanonas/uso terapêutico , Humanos , Melanoma Experimental/patologia , Camundongos , Neovascularização Patológica/patologia , Ratos , Neoplasias Cutâneas/patologia
8.
Int Immunopharmacol ; 89(Pt A): 107046, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33045572

RESUMO

Diosmetin, a citrus flavonoid, has a variety of therapeutic properties such as antibacterial, anti-inflammatory and antioxidant effects. However, the effect of diosmetin on atopic dermatitis (AD) development has not been reported. This study thus aims to investigate whether diosmetin possesses inhibitory effects on AD development. A dinitrochlorobenzene (DNCB)-induced AD mouse model was used to evaluate the effects of diosmetin on AD development. Treatment with diosmetin significantly reduced the dermatitis score, thickness of epidermis and dermis and number of mast cells in comparison with the untreated group. Furthermore, immunohistochemical analysis using an anti-F4/80 antibody demonstrated that diosmetin significantly suppressed macrophage infiltration into the AD lesion. It was observed that the levels of pro-inflammatory cytokines (TNF-α, IL-4 and IL-1ß) in skin lesion decreased in response to treatment with diosmetin. In addition, the anti-inflammatory effect of diosmetin was evaluated in LPS- or IL-4-induced a mouse macrophage cell line (raw 264.7). Diosmetin inhibited the production of nitric oxide and decreased the expression of inducible nitric oxide synthase (iNOS). Diosmetin not only suppressed the phosphorylation of MAP kinase (ERK 1/2, p38 and JNK) but the activation of JAK/STAT signaling. The mRNA analysis demonstrated that diosmetin also reduced the level of inflammatory cytokines such as IL-1ß and IL-6. Collectively, these results demonstrate that diosmetin exhibits the inhibitory effect on AD, suggesting that diosmetin may be a potential therapeutic agent for this atopic disorder.


Assuntos
Anti-Inflamatórios/farmacologia , Dermatite Atópica/induzido quimicamente , Flavonoides/farmacologia , Interleucina-4/toxicidade , Lipopolissacarídeos/toxicidade , Animais , Anti-Inflamatórios/química , Dermatite Atópica/tratamento farmacológico , Dinitroclorobenzeno/toxicidade , Flavonoides/administração & dosagem , Ativação de Macrófagos , Camundongos , Células RAW 264.7
9.
Mol Med Rep ; 22(2): 1235-1242, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32468067

RESUMO

During pregnancy, the uterus undergoes intense neovascularization and vascular remodeling to supply oxygen and nutrients to the embryo. During this period, progesterone secreted from the ovary has effects on vascular remodeling in the endometrium and interacts with angiogenic factors. However, the exact mechanism of uterine vascular remodeling during pregnancy is poorly understood. Therefore, the aim of the present study was to investigate the association between angiopoietin-2 (Ang-2), one of the angiopoietins, and intrauterine vessel remodeling during pregnancy, and to determine the effect of progesterone on Ang-2 levels. Changes in Ang-2 expression were observed according to quantitative modification of progesterone using pregnant mice and human uterine microvascular endothelial cells. As a result, Ang-2 was observed mainly in the mesometrial region (MR) of the uterus during the period between implantation and placentation. Furthermore, a substantial amount of Ang-2 also appeared in endothelial cells, particularly of the venous sinus region (VSR). Interestingly, Ang-2 expression was increased by progesterone, whereas estrogen had limited effects. To confirm the association between Ang-2 and progesterone, the function of the progesterone receptor (PR) was inhibited using RU486, a blocker of PR. Ang-2 expression and vascular remodeling of the VSR in the uterus were decreased when the functions of progesterone were inhibited. Overall, the regulation of Ang-2 by progesterone/PR was associated with vascular remodeling in the VSR during pregnancy. The present study proposed a solution to prevent pregnancy failure due to a lack of vascularity in the uterus in advance.


Assuntos
Angiopoietina-2/metabolismo , Neovascularização Fisiológica/fisiologia , Progesterona/farmacologia , Útero/irrigação sanguínea , Útero/metabolismo , Remodelação Vascular/fisiologia , Animais , Células Cultivadas , Implantação do Embrião/fisiologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/crescimento & desenvolvimento , Endotélio Vascular/metabolismo , Feminino , Antagonistas de Hormônios/farmacologia , Humanos , Camundongos Endogâmicos C57BL , Mifepristona/farmacologia , Gravidez , Receptores de Progesterona/antagonistas & inibidores , Receptores de Progesterona/metabolismo , Análise Espaço-Temporal , Útero/citologia , Remodelação Vascular/efeitos dos fármacos
10.
Biomed Pharmacother ; 117: 109091, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31228803

RESUMO

Diosmetin is a natural flavonoid obtained from citrus fruits and some medicinal herbs. Previous studies have reported the anti-cancer activity of diosmetin in some types of tumors. However, it is still unclear whether diosmetin exerts anti-cancer effects, particularly anti-angiogenic effects, in skin cancer. In this study, we used B16F10 melanoma cells and human umbilical vein endothelial cells to investigate the inhibitory effect of diosmetin on cell proliferation, migration and tube formation in vitro. Rat aorta ring assays were performed to determine the effect of diosmetin on ECs sprouting ex vivo. Furthermore, a B16F10 mouse melanoma model was used to observe the effect of diosmetin on tumor growth, angiogenesis, and metastasis in vivo. Our results showed that diosmetin not only suppressed tumor cell proliferation and migration but also induced cell apoptosis via the caspase pathway in B16F10 cells, and potently inhibited tube formation and cell migration in HUVECs. Rat aorta ring assays showed that diosmetin attenuated the ECs sprouting. Moreover, the mouse melanoma model showed that diosmetin significantly delayed tumor growth by inhibiting tumor vessels sprouting and expansion during tumor progression. Notably, diosmetin induced the normalization of tumor vasculature through the downregulation of angiopoietin-2 and the improvement of pericyte coverage, leading to suppression of metastasis formation in lungs and lymph nodes. In conclusion, our results demonstrate that diosmetin suppresses tumor progression and metastasis by inducing tumor cell death and inhibiting tumor angiogenesis as well as normalizing the defective tumor vasculature, suggesting that diosmetin is a potential adjuvant chemotherapy agent.


Assuntos
Inibidores da Angiogênese/farmacologia , Antineoplásicos/farmacologia , Flavonoides/farmacologia , Neovascularização Patológica/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana , Humanos , Metástase Linfática/tratamento farmacológico , Metástase Linfática/patologia , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/patologia , Ratos , Ratos Sprague-Dawley , Neoplasias Cutâneas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
11.
Biomed Pharmacother ; 106: 326-332, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29966977

RESUMO

Alchemilla vulgaris and Mimosa tenuiflora (Mimosa) have been used to treat cutaneous wounds as a traditional remedy due to their various biological activities. But, there are only a few studies about the effects of these herbs on wound healing. The purpose of this study is to investigate the wound healing effect of the herbal mixture, consisting of A. vulgaris and Mimosa, in mice and to determine the activity of the extract in vitro. In present study, application of an ointment containing the herbal mixture on the dorsal skin wounds of mice showed that the wound healing process was faster than treatment of Fusidic acid. Histological analysis demonstrated the herbal mixture promoted re-epithelialization, collagen synthesis, and especially the regeneration of skin appendages such as hair follicles. Immunohistochemical analysis revealed the herbal mixture improved angiogenesis and the stabilization of blood vessels, as well as accelerated the formation of granulation tissue. In addition, we demonstrated that herbal mixture enhanced the migration of HaCaT, fibroblasts, and HUVECs on a two-dimensional wound, and promoted the proliferation of macrophages and lymphatic vessels. Our results demonstrated that herbal mixture can promote the migration of keratinocytes, fibroblasts, and endothelial cells, and the proliferation of macrophages and lymphatic vessels. Furthermore, it showed that herbal mixture accelerates wound healing. Therefore, we suggest that herbal mixture may have a potential for therapeutic use for treatment and management of cutaneous wound.


Assuntos
Alchemilla , Fármacos Dermatológicos/farmacologia , Extratos Vegetais/farmacologia , Pele/efeitos dos fármacos , Cicatrização/efeitos dos fármacos , Ferimentos Penetrantes/tratamento farmacológico , Células 3T3-L1 , Administração Cutânea , Alchemilla/química , Animais , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Colágeno/metabolismo , Fármacos Dermatológicos/administração & dosagem , Fármacos Dermatológicos/isolamento & purificação , Modelos Animais de Doenças , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Queratinócitos/patologia , Vasos Linfáticos/efeitos dos fármacos , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Mimosa/química , Neovascularização Fisiológica/efeitos dos fármacos , Pomadas , Fitoterapia , Extratos Vegetais/administração & dosagem , Extratos Vegetais/isolamento & purificação , Plantas Medicinais , Reepitelização/efeitos dos fármacos , Pele/lesões , Pele/metabolismo , Pele/patologia , Fatores de Tempo , Ferimentos Penetrantes/metabolismo , Ferimentos Penetrantes/patologia
12.
Oncol Rep ; 38(5): 3011-3018, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29048641

RESUMO

Baicalein, a herbal medicine, is a natural flavonoid isolated from the roots of Scutellaria baicalensis Georgi. It is known for its anticancer, anti-inflammatory and neuroprotective properties. Despite these well-known properties, it is not yet clear what effect baicalein has on tumor progression. Therefore, in the present study, we used B16F10 cells, Lewis lung carcinoma (LLC) cells, and human umbilical vein endothelial cells (HUVECs) to investigate the effect of baicalein on cell proliferation and viability, migration and tube formation in vitro. In addition, an experimental animal model was used to observe the growth rate and metastasis of tumors and tumor vessel formation in vivo. Our results showed that baicalein decreased the proliferation and migration and induced tumor cell death via caspase-3 activation in the B16F10 and LLC cells, and strongly inhibited tube formation and cell migration in HUVECs. Furthermore, mouse models showed that baicalein reduced the tumor volume and greatly reduced the tumor growth rate in the early stages of tumor progression, and the baicalein-treated groups had significantly reduced expression of CD31 (endothelial cell marker) and α-SMA (mural cell marker) in the tumors, indicating that baicalein inhibits tumor angiogenesis by disrupting tumor vasculature development. Comparison of the lymph node and lung samples collected from the baicalein-treated group, and the untreated group showed that baicalein reduced metastasis of the tumor to these tissues. In summary, baicalein reduced tumor progression and metastasis, directly induced tumor cell death, and inhibited tumor angiogenesis. Our results strongly demonstrate that baicalein is a potential chemotherapeutic agent.


Assuntos
Proliferação de Células/efeitos dos fármacos , Flavanonas/administração & dosagem , Melanoma Experimental/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Extratos Vegetais/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Lewis , Movimento Celular/efeitos dos fármacos , Flavanonas/química , Células Endoteliais da Veia Umbilical Humana , Humanos , Melanoma Experimental/genética , Melanoma Experimental/patologia , Camundongos , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Extratos Vegetais/química , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Scutellaria baicalensis
13.
Int J Mol Med ; 40(4): 1210-1216, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28849193

RESUMO

Early pregnancy is characterized by an increase in the blood volume of the uterus for embryonic development, thereby exerting fluid shear stress (FSS) on the vascular walls. The uterus experiences vascular remodeling to accommodate the increased blood flow. The blood flow­induced FSS elevates the expression of vascular endothelial growth factors (VEGFs) and their receptors, and regulates vascular remodeling through the activation of VEGF receptor-3 (VEGFR-3). However, the mechanisms responsible for FSS-induced VEGFR-3 expression in the uterus during pregnancy are unclear. In this study, we demonstrate that vascular remodeling in the uterus during pregnancy is regulated by FSS-induced VEGFR-3 expression. We examined the association between VEGFR-3 and FSS through in vivo and in vitro experiments. In vivo experiments revealed VEGFR-3 expression in the CD31-positive region of the uterus of pregnant mice; VEGF-C (ligand for VEGFR­3) was undetected in the uterus. These results confirmed that VEGFR-3 expression in the endometrium is independent of its ligand. In vitro studies experiments revealed that FSS induced morphological changes and increased VEGFR-3 expression in human uterine microvascular endothelial cells. Thus, VEGFR-3 activation by FSS is associated with vascular remodeling to allow increased blood flow in the uterus during pregnancy.


Assuntos
Endotélio Vascular/metabolismo , Mecanotransdução Celular , Útero/metabolismo , Fator C de Crescimento do Endotélio Vascular/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética , Animais , Embrião de Mamíferos , Desenvolvimento Embrionário/fisiologia , Endotélio Vascular/crescimento & desenvolvimento , Feminino , Regulação da Expressão Gênica , Humanos , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Reologia , Estresse Mecânico , Útero/irrigação sanguínea , Útero/crescimento & desenvolvimento , Fator C de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Remodelação Vascular/fisiologia
14.
Int J Biol Macromol ; 76: 45-8, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25709023

RESUMO

Human hair-based biocomposite nanofibers (NFs) have been fabricated by an electrospinning technique. Aqueous keratin extracted from human hair was successfully blended with poly(vinyl alcohol) (PVA). The focus here is on transforming into keratin/PVA nanofibrous membranes and insoluble property of electrospun NFs. The resulting hair-based NFs were characterized using Fourier transform infrared (FT-IR) spectroscopy, scanning electron microscopy (SEM), X-ray diffraction (XRD), differential scanning colorimetry (DSC), and thermogravimetric analysis (TGA). Toward the potential use of these NFs after cross-linking with various weight fractions of glyoxal, its physicochemical properties, such as morphology, mechanical strength, crystallinity, and chemical structure were investigated. Keratin/PVA ratio of 2/1 NFs with 6 wt%-glyoxal showed good uniformity in fiber morphology and suitable mechanical properties, and excellent antibacterial activity providing a potential application of hair-based NFs in biomedical field.


Assuntos
Cabelo/química , Nanofibras/química , Antibacterianos/química , Antibacterianos/farmacologia , Testes de Sensibilidade a Antimicrobianos por Disco-Difusão , Cabelo/ultraestrutura , Humanos , Queratinas/química , Nanofibras/ultraestrutura , Álcool de Polivinil/química , Espectroscopia de Infravermelho com Transformada de Fourier , Difração de Raios X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA