Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 68
1.
Int J Mol Sci ; 25(5)2024 Feb 20.
Article En | MEDLINE | ID: mdl-38473712

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.


Mammary Glands, Human , Oxazoles , Receptors, Estrogen , Dogs , Animals , Female , Humans , Receptors, Estrogen/metabolism , Genistein/pharmacology , Estrogen Receptor beta/genetics , Estrogen Receptor alpha/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Down-Regulation , Mammary Glands, Human/metabolism , Estrogens/metabolism
2.
IBRO Neurosci Rep ; 16: 336-344, 2024 Jun.
Article En | MEDLINE | ID: mdl-38390232

Alzheimer's disease (AD) is the most common age-related progressive neurodegenerative disorder. The accumulation of amyloid beta-peptide is a neuropathological marker of AD. While melatonin is recognized to have protective effects on aging and neurodegenerative disorders, the therapeutic effect of melatonin on calcineurin in AD is poorly understood. In this study, we examined the effect and underlying molecular mechanisms of melatonin treatment on amyloid beta-mediated neurotoxicity in neuroblastoma cells. Melatonin treatment decreased calcineurin and autophagy in neuroblastoma cells. Electron microscopy images showed that melatonin inhibited amyloid beta-induced autophagic vacuoles. The increase in the amyloid beta-induced apoptosis rate was observed more in PrPC-expressing ZW cells than in PrPC-silencing Zpl cells. Taken together, the results suggest that by mitigating the effect of calcineurin and autophagy flux activation, melatonin could also rescue amyloid beta-induced neurotoxic effects. These findings may be relevant to therapy for neurodegenerative diseases, including AD.

3.
BMC Neurosci ; 24(1): 59, 2023 11 06.
Article En | MEDLINE | ID: mdl-37932682

BACKGROUND: Japanese encephalitis virus (JEV) is a mosquito-borne flavivirus that has no specific treatment except for supportive medical care. JEV is a neurotropic virus that affects the nervous system and triggers inflammation in the brain. METHODS: Melatonin is used as a sleep-inducing agent in neurophysiology and may serve as a protective agent against neurological and neurodegenerative diseases. Herein, we investigated the effects of melatonin and the critical roles of the serine/threonine protein phosphatase calcineurin during JEV infection in SK-N-SH neuroblastoma cells. RESULTS: Melatonin treatment decreased JEV replication and JEV-mediated neurotoxicity. Calcineurin activity was increased by JEV infection and inhibited by melatonin treatment. Through calcineurin regulation, melatonin decreased the JEV-mediated neuroinflammatory response and attenuated JEV-induced autophagy. CONCLUSIONS: Calcineurin inactivation has a protective effect in JEV-infected neuronal cells, and melatonin is a novel resource for the development of anti-JEV agents.


Encephalitis Virus, Japanese , Encephalitis, Japanese , Melatonin , Animals , Humans , Encephalitis Virus, Japanese/physiology , Calcineurin/pharmacology , Melatonin/pharmacology , Autophagy
4.
BMC Vet Res ; 19(1): 223, 2023 Oct 25.
Article En | MEDLINE | ID: mdl-37880653

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.


Mammary Glands, Human , Phosphatidylinositol 3-Kinases , Dogs , Animals , Female , Mice , Humans , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/pharmacology , Endothelial Cells/metabolism , Mammary Glands, Human/metabolism , Cell Line, Tumor , Cell Proliferation
5.
Virus Res ; 338: 199249, 2023 12.
Article En | MEDLINE | ID: mdl-37858731

Flaviviruses are a major cause of viral diseases worldwide, for which effective treatments have yet to be discovered. The prion protein (PrPc) is abundantly expressed in brain cells and has been shown to play a variety of roles, including neuroprotection, cell homeostasis, and regulation of cellular signaling. However, it is still unclear whether PrPc can protect against flaviviruses. In this study, we investigated the role of PrPc in regulating autophagy flux and its potential antiviral activity during Japanese encephalitis virus (JEV) infection. Our in vivo experiment showed that JEV was more lethal to the PrPc knocked out mice which was further supported by histological analysis, western blot and rtPCR results from infected mice brain samples. Role of PrPc against viral propagation in vitro was verified through cell survival study, protein expression and RNA replication analysis, and adenoviral vector assay by overexpressing PrPc. Further analysis indicated that after virus entry, PrPc inhibited autophagic flux that prevented JEV replication inside the host cell. Our results from in vivo and in vitro investigations demonstrate that prion protein effectively inhibited JEV propagation by regulating autophagy flux which is used by JEV to release its genetic material and replication after entering the host cell, suggesting that prion protein may be a promising therapeutic target for flavivirus infection.


Encephalitis Virus, Japanese , Encephalitis, Japanese , Animals , Mice , Prion Proteins/genetics , Prion Proteins/pharmacology , Cell Line , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Virus Replication
6.
Anticancer Agents Med Chem ; 23(20): 2225-2236, 2023.
Article En | MEDLINE | ID: mdl-37859313

BACKGROUND: TRAIL has emerged as a promising therapeutic target due to its ability to selectively induce apoptosis in cancer cells while sparing normal cells. Autophagy, a highly regulated cellular recycling mechanism, is known to play a cell survival role by providing a required environment for the cell. Recent studies suggest that autophagy plays a significant role in increasing TRAIL resistance in certain cancer cells. Thus, regulating autophagy in TRAIL-mediated cancer therapy is crucial for its role in cancer treatment. OBJECTIVE: Our study explored whether the antidepressant drug desipramine could enhance the ability of TRAIL to kill cancer cells by inhibiting autophagy. METHODS: The effect of desipramine on TRAIL sensitivity was examined in various lung cancer cell lines. Cell viability was measured by morphological analysis, trypan blue exclusion, and crystal violet staining. Flow cytometry analysis was carried out to measure apoptosis with annexin V-PI stained cells. Western blotting, rtPCR, and immunocytochemistry were carried out to measure autophagy and death receptor expression. TEM was carried out to detect autophagy inhibition. RESULTS: Desipramine treatment increased the TRAIL sensitivity in all lung cancer cell lines. Mechanistically, desipramine treatment induced death receptor expression to increase TRAIL sensitivity. This effect was confirmed when the genetic blockade of DR5 reduced the effect of desipramine in enhanced TRAIL-mediated cell death. Further investigation revealed that desipramine treatment increased the LC3 and p62 levels, indicating the inhibition of lysosomal degradation of autophagy. Notably, TRAIL, in combination with either desipramine or the autophagy inhibitor chloroquine, exhibited enhanced cytotoxicity compared to TRAIL treatment alone. CONCLUSION: Our findings revealed the potential of desipramine to induce TRAIL-mediated cell death by autophagy impairment. This discovery suggests its therapeutic potential for inducing TRAIL-mediated cell death by increasing the expression of death receptors, which is caused by impairing autophagy.


Desipramine , Lung Neoplasms , Receptors, TNF-Related Apoptosis-Inducing Ligand , Humans , Antidepressive Agents/pharmacology , Apoptosis/drug effects , Autophagy , Cell Line, Tumor , Desipramine/pharmacology , Desipramine/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/drug effects , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology
7.
Int J Vet Sci Med ; 11(1): 87-93, 2023.
Article En | MEDLINE | ID: mdl-37649734

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.

8.
Vet Sci ; 10(2)2023 Jan 22.
Article En | MEDLINE | ID: mdl-36851388

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.

9.
Front Vet Sci ; 9: 1003351, 2022.
Article En | MEDLINE | ID: mdl-36246324

The past and current burden of swine influenza A viruses (swIAV) must be estimated since pigs act as mixing vessels and are considered a potential source of newly emerging IAV variants. The objective of this systematic review and meta-analysis was to integrate data on the prevalence and seroprevalence of swIAV in South Korean domestic pigs and evaluate important risk factors that influence these outcomes. Eight databases were searched for studies that evaluated the prevalence and seroprevalence of swIAV in South Korean pigs using a specified search string; twenty-seven eligible studies were identified after application of a set of pre-determined inclusion criteria by three authors. The reported prevalence and seroprevalence were pooled separately in proportions between 0 and 1, using a random-effect meta-analysis. To identify and quantify potential sources of heterogeneity, subgroup, and meta-regression analyses were conducted using covariates (publication type, swIAV subtype, growth stage of pigs, sampling region, publication year, sampling season, facility, detection method, sample type, and sample size). The overall prevalence and seroprevalence in domestic pigs were 0.05 [95% confidence intervals (CIs): 0.05-0.12] and 0.35 (95% CIs: 0.14-0.63), respectively. To identify the impact of covariates on effect size, a suitable meta-regression model was determined using predictor importance estimates with corrected Akaike information criterion values. Consequently, the best-fit model included two covariates, publication year and sample size, which were significantly associated with high heterogeneity in the subgroup analysis. Furthermore, data visualization depicted a significant non-linear association between swIAV prevalence and seroprevalence and specific growth stages of pigs. These findings suggest that the periodic monitoring of pigs at different growth stages in large farms may help to establish the status of swIAV-spread across species in the region, and thereby minimize pandemic risk.

10.
Article En | MEDLINE | ID: mdl-36110191

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.

11.
Animals (Basel) ; 11(6)2021 May 23.
Article En | MEDLINE | ID: mdl-34071074

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.

12.
Int J Med Sci ; 17(18): 3049-3057, 2020.
Article En | MEDLINE | ID: mdl-33173425

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.


Angiogenesis Inhibitors/pharmacology , Flavanones/pharmacology , Melanoma, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Skin Neoplasms/drug therapy , Angiogenesis Inhibitors/therapeutic use , Animals , Apoptosis/drug effects , Cell Line, Tumor , Flavanones/therapeutic use , Humans , Melanoma, Experimental/pathology , Mice , Neovascularization, Pathologic/pathology , Rats , Skin Neoplasms/pathology
13.
Int Immunopharmacol ; 89(Pt A): 107046, 2020 Dec.
Article En | MEDLINE | ID: mdl-33045572

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.


Anti-Inflammatory Agents/pharmacology , Dermatitis, Atopic/chemically induced , Flavonoids/pharmacology , Interleukin-4/toxicity , Lipopolysaccharides/toxicity , Animals , Anti-Inflammatory Agents/chemistry , Dermatitis, Atopic/drug therapy , Dinitrochlorobenzene/toxicity , Flavonoids/administration & dosage , Macrophage Activation , Mice , RAW 264.7 Cells
14.
Int J Mol Med ; 46(2): 795-805, 2020 Aug.
Article En | MEDLINE | ID: mdl-32626921

Tumor necrosis factor­related apoptosis­inducing ligand (TRAIL) is a potential target for cancer therapy, owing to its ability to selectively kill cancer cells without causing significant toxicity to normal cells. However, due to the lack of death receptor expression, cancer cells can become highly resistant to TRAIL. Hence, it is vital to develop agents that restore TRAIL efficacy. Sertraline is an antidepressant drug with anticancer properties. To the best of our knowledge, this is the first study to demonstrate that sertraline inhibits autophagic flux and increases the expression of death receptor 5 (DR5) on TRAIL­resistant lung cancer cells. Inhibition of autophagy using autophagy inhibitors 3­methyladenine and chloroquine upregulated the expression of DR5 and enhanced TRAIL­induced apoptosis, as confirmed by the increase of pro­apoptotic proteins caspase­8 and caspase­3. Silencing DR5 expression using DR5 small interfering RNA prevented sertraline­induced TRAIL­mediated apoptosis, indicating the role of DR5 in TRAIL­mediated apoptosis. Overall, sertraline enhanced TRAIL­mediated apoptosis via the downregulation of AMP­activated protein kinase phosphorylation, resulting in the inhibition of autophagic flux, upregulation of DR5 expression, and activation of the apoptotic caspase cascade. These data suggested that sertraline could be used to sensitize human lung cancer cells to TRAIL, while also serving as a therapeutic option in cancer patients with depression.


Lung Neoplasms/drug therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Sertraline/therapeutic use , TNF-Related Apoptosis-Inducing Ligand/pharmacology , A549 Cells , Apoptosis/drug effects , Autophagy/drug effects , Autophagy/genetics , Blotting, Western , Caspase 3/genetics , Caspase 3/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Cell Survival/drug effects , Cell Survival/genetics , Humans , Immunohistochemistry , Lung Neoplasms/metabolism , Microscopy, Electron, Transmission , Phosphorylation/drug effects , Phosphorylation/genetics , RNA Interference , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics
15.
Mol Med Rep ; 22(2): 1235-1242, 2020 08.
Article En | MEDLINE | ID: mdl-32468067

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.


Angiopoietin-2/metabolism , Neovascularization, Physiologic/physiology , Progesterone/pharmacology , Uterus/blood supply , Uterus/metabolism , Vascular Remodeling/physiology , Animals , Cells, Cultured , Embryo Implantation/physiology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/growth & development , Endothelium, Vascular/metabolism , Female , Hormone Antagonists/pharmacology , Humans , Mice, Inbred C57BL , Mifepristone/pharmacology , Pregnancy , Receptors, Progesterone/antagonists & inhibitors , Receptors, Progesterone/metabolism , Spatio-Temporal Analysis , Uterus/cytology , Vascular Remodeling/drug effects
16.
Biomed Pharmacother ; 117: 109091, 2019 Sep.
Article En | MEDLINE | ID: mdl-31228803

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.


Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Flavonoids/pharmacology , Neovascularization, Pathologic/drug therapy , Skin Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Lymphatic Metastasis/drug therapy , Lymphatic Metastasis/pathology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/pathology , Rats , Rats, Sprague-Dawley , Skin Neoplasms/pathology , Xenograft Model Antitumor Assays/methods
17.
Mol Neurobiol ; 56(6): 4192-4202, 2019 Jun.
Article En | MEDLINE | ID: mdl-30288697

Accumulation of prion protein (PrPc) into a protease-resistant form (PrPsc) in the brains of humans and animals affects the central nervous system. PrPsc occurs only in mammals with transmissible prion diseases. Prion protein refers to either the infectious pathogen itself or the main component of the pathogen. Recent studies suggest that autophagy is one of the major functions that keep cells alive and which has a protective effect against neurodegeneration. In this study, we investigated whether the anti-hypertensive drug, captopril, could attenuate prion peptide PrP (106-126)-induced calcium alteration-mediated neurotoxicity. Treatment with captopril increased both LC3-II (microtubule-associated protein 1A/1B-light chain 3-II) and p62 protein levels, indicating autophagy flux inhibition. Electron microscopy confirmed the occurrence of autophagic flux inhibition in neuronal cells treated with captopril. Captopril attenuated PrP (106-126)-induced neuronal cell death via AMPK activation and autophagy inhibition. Compound C suppressed AMPK activation as well as the neuroprotective effects of captopril. Thus, these data showed that an anti-hypertensive drug has a protective effect against prion-mediated neuronal cell death via autophagy inhibition and AMPK activation, and also suggest that anti-hypertensive drugs may be effective therapeutic agents against neurodegenerative disorders, including prion diseases.


AMP-Activated Protein Kinases/metabolism , Apoptosis/drug effects , Autophagy/drug effects , Captopril/pharmacology , Neurons/enzymology , Neurons/pathology , Peptides/metabolism , Prions/metabolism , Animals , Calcium/metabolism , Enzyme Activation/drug effects , Mice , Neurons/drug effects , Neurons/ultrastructure , Neurotoxins/toxicity
18.
Mol Cell Endocrinol ; 481: 84-94, 2019 02 05.
Article En | MEDLINE | ID: mdl-30513342

The role of autophagy modulation in adipogenic differentiation and the possible autophagy modulators targeting adipogenesis remain unclear. In this study, we investigated whether normal cellular prion protein (PrP) is involved in the modulation of autophagy and affects adipogenic differentiation in vivo and in vitro. Surprisingly, autophagy flux signals were activated in the adipose tissue of prion protein-deficient mice and PrP-deleted 3T3-L1 adipocytes. The activation of autophagy flux mediated by PrP deletion was confirmed in the adipose tissue via transmission electron microscopy. Adipocyte differentiation factors were highly induced in prion protein-deficient adipose tissue and 3T3-L1 adipocytes. In addition, deletion of prion protein significantly increased visceral fat volume, body fat weight, adipocyte cell size, and body weight gain in Prnp-knockout mice and increased lipid accumulation in PrP siRNA-transfected 3T3-L1 cells. However, the overexpression of prion protein using adenovirus inhibited the autophagic flux signals, lipid accumulation, and the PPAR-γ and C/EBP-α mRNA and protein expression levels in comparison to those in the control cells. Our results demonstrated that deletion of normal prion protein accelerated adipogenic differentiation and lipid accumulation mediated via autophagy flux activation.


Adipocytes/cytology , Intra-Abdominal Fat/metabolism , Prion Proteins/genetics , Prion Proteins/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Autophagy , Body Weight , Cell Differentiation , Gene Deletion , Mice
19.
20.
Biomed Pharmacother ; 106: 326-332, 2018 Oct.
Article En | MEDLINE | ID: mdl-29966977

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.


Alchemilla , Dermatologic Agents/pharmacology , Plant Extracts/pharmacology , Skin/drug effects , Wound Healing/drug effects , Wounds, Penetrating/drug therapy , 3T3-L1 Cells , Administration, Cutaneous , Alchemilla/chemistry , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Collagen/metabolism , Dermatologic Agents/administration & dosage , Dermatologic Agents/isolation & purification , Disease Models, Animal , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/pathology , Lymphatic Vessels/drug effects , Lymphatic Vessels/metabolism , Lymphatic Vessels/pathology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred BALB C , Mimosa/chemistry , Neovascularization, Physiologic/drug effects , Ointments , Phytotherapy , Plant Extracts/administration & dosage , Plant Extracts/isolation & purification , Plants, Medicinal , Re-Epithelialization/drug effects , Skin/injuries , Skin/metabolism , Skin/pathology , Time Factors , Wounds, Penetrating/metabolism , Wounds, Penetrating/pathology
...