Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
NPJ Aging ; 9(1): 11, 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37264012

ABSTRACT

Vascular aging is a major contributing factor to cardiovascular disease. The aged blood vessels, characterized by vascular wall thickening and stiffening, are instigated by endothelial cell dysfunction induced by oxidative stress and inflammation. von Willebrand Factor (vWF) is a glycoprotein known for its role in coagulation, and plasma levels of vWF are increased with age. Elevated vWF promotes thrombosis, atherosclerotic plaque formation, inflammation and proliferation of vascular smooth muscle cells. Cadmium (Cd) is an environmental pollutant associated with increased morbidity and mortality of cardiovascular disease. At low concentrations, Cd activates pro-survival signaling in endothelial cells, however enhances intima-media thickness and atherogenesis. A non-cytotoxic dose of Cd also increases endothelial vWF expression and secretion in vivo and in vitro. In this review, we summarize the molecular mechanisms underlying vWF-promoted vascular aging-associated pathologies and Cd-induced vWF expression. In addition, we propose that exposure to low-dose Cd is a risk factor for vascular aging, through elevation of plasma vWF.

2.
Article in English | MEDLINE | ID: mdl-36497941

ABSTRACT

Osteoporosis (OP) is a degenerative disease characterized by reduced bone strength and increased fracture risk. As the global population continues to age, the prevalence and economic burden of osteoporosis can be expected to rise substantially, but there remain various gaps in the field of OP care. For instance, there is a lack of anti-fracture drugs with proven long-term efficacy. Likewise, though exercise remains widely recommended in OP prevention and management, data regarding the safety and efficacy for patients after vertebral fracture remain limited. This lack of evidence may be due to the cost and inherent difficulties associated with exercise-based OP research. Thus, the current research landscape highlights the need for novel research strategies that accelerate OP drug discovery and allow for the low-cost study of exercise interventions. Here, we outline an example of one strategy, the use of zebrafish, which has emerged as a potential model for the discovery of anti-osteoporosis therapeutics and study of exercise interventions. The strengths, limitations, and potential applications of zebrafish in OP research will be outlined.


Subject(s)
Fractures, Bone , Osteoporosis , Animals , Zebrafish , Bone Density , Osteoporosis/drug therapy , Osteoporosis/complications
3.
Birth Defects Res ; 113(20): 1427-1430, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34800008
4.
Aging (Albany NY) ; 11(16): 6602-6613, 2019 08 28.
Article in English | MEDLINE | ID: mdl-31461408

ABSTRACT

Lymphatic vessels maintain body homeostasis by recirculation of fluid and cells. Cell senescence induces lymphatic dysfunction. Impaired contractile function is caused by low muscle cell investiture and decrease of nitric oxide in aged lymphatic collectors, leading to poor drainage of lymph. Aging-induced loss of endothelial glycocalyx and production of inflammatory cytokines increases permeability of lymphatic vessels. In addition, aging-associated basal activation of mast cells delays immune response. In this review, we summarize the structural and pathological changes of aged lymphatic vessels, and discuss the underlying molecular mechanisms.


Subject(s)
Aging/pathology , Lymphatic Vessels/physiopathology , Humans
5.
J Immunol ; 202(4): 1210-1218, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30642982

ABSTRACT

Cadmium (Cd) is an environmental toxin that induces nephrotoxicity. Complement factor H (CFH), an inhibitor of complement activation, is involved in the pathogenesis of various renal diseases. In this study, we investigated the effects of Cd on CFH production by the kidney. In C57B6/J mice, an increased CFH level was found in renal blood and glomerular endothelial cells after Cd treatment. In vitro, Cd induces an increased CFH secretion and mRNA expression in human renal glomerular endothelial cells but not in human podocytes or human mesangial cells. Cd activates the JNK pathway and increases c-Jun and c-Fos in human renal glomerular endothelial cells. A JNK inhibitor, SP600125, specifically abolishes Cd-induced CFH production. By chromatin immunoprecipitation assay and EMSA, the -1635 AP-1 motif on human CFH promoter was identified as the binding element for c-Jun and c-Fos. In a luciferase activity assay, mutation of the AP1 site eliminates Cd-induced increase of CFH promoter activity. Thus, the -1635 AP-1 motif on the CFH promoter region mediates Cd-inducible CFH gene expression.


Subject(s)
Cadmium/pharmacology , Complement Factor H/metabolism , Endothelial Cells/drug effects , Kidney Glomerulus/drug effects , Transcription Factor AP-1/metabolism , Animals , Anthracenes/pharmacology , Binding Sites/drug effects , Cells, Cultured , Complement Factor H/antagonists & inhibitors , Complement Factor H/genetics , Endothelial Cells/metabolism , Humans , Kidney Glomerulus/metabolism , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factor AP-1/antagonists & inhibitors , Transcription Factor AP-1/genetics
6.
Toxicol Lett ; 290: 46-54, 2018 Jun 15.
Article in English | MEDLINE | ID: mdl-29571895

ABSTRACT

Cadmium (Cd) is a persistent and widespread environmental pollutant of continuing worldwide concern. Previous studies have suggested that Cd exposure increases the risk of cardiovascular diseases, such as atherosclerosis and hypertension. However, the underlying mechanisms are poorly understood. In this study, we observed that low dose Cd treatment induced von Willebrand factor (vWF) expression in vascular endothelial cells in mouse lung and kidney tissues. In vitro analysis showed that 1 µM Cd specifically upregulated vWF mRNA and protein expression in human umbilical vein endothelial cells (HUVECs), indicating that Cd targets vascular endothelial cells even at relatively low concentrations. Further study demonstrated that nuclear factor kappa B (NF-κB) and GATA3, two established transcription regulators of the vWF gene, were not altered in the presence of Cd. However, ETS-related gene (ERG) was significantly induced by 1 µM Cd. When ERG was knocked down by siRNA, Cd induced upregulation of vWF was totally blocked. Chromatin immunoprecipitation (ChIP) assay showed that Cd increases the binding of ERG on the -56 ETS motif on the human vWF promoter. These results indicated that ERG mediated the increased expression of vWF by Cd. Since vWF is a key regulator for vascular homeostasis, our findings may provide a novel mechanism for understanding low dose Cd induced development of vascular diseases.


Subject(s)
Cadmium/toxicity , Endothelial Cells/drug effects , von Willebrand Factor/genetics , Animals , Cells, Cultured , Endothelial Cells/metabolism , GATA3 Transcription Factor/genetics , Humans , Male , Mice , NF-kappa B/physiology , Oncogene Proteins/genetics , Signal Transduction/drug effects , Transcriptional Regulator ERG/genetics , Up-Regulation
7.
Theranostics ; 8(21): 6053-6069, 2018.
Article in English | MEDLINE | ID: mdl-30613282

ABSTRACT

Identified as a major downstream effector of the small GTPase RhoA, Rho-associated coiled-coil kinase (ROCK) is a versatile regulator of multiple cellular processes. Angiogenesis, the process of generating new capillaries from the pre-existing ones, is required for the development of various diseases such as cancer, diabetes and rheumatoid arthritis. Recently, ROCK has attracted attention for its crucial role in angiogenesis, making it a promising target for new therapeutic approaches. In this review, we summarize recent advances in understanding the role of ROCK signaling in regulating the permeability, migration, proliferation and tubulogenesis of endothelial cells (ECs), as well as its functions in non-ECs which constitute the pro-angiogenic microenvironment. The therapeutic potential of ROCK inhibitors in angiogenesis-related diseases is also discussed.


Subject(s)
Gene Expression Regulation , Neovascularization, Pathologic , Neovascularization, Physiologic , rho-Associated Kinases/metabolism , Animals , Endothelial Cells/physiology , Humans
8.
Oncotarget ; 8(40): 69139-69161, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28978186

ABSTRACT

Angiogenesis is the development of new blood vessels, which is required for tumor growth and metastasis. Signal transducer and activator of transcription factor 3 (STAT3) is a transcription factor that regulates a variety of cellular events including proliferation, differentiation and apoptosis. Previous studies revealed that activation of STAT3 promotes tumor angiogenesis. In this review, we described the activities of STAT3 signaling in different cell types involved in angiogenesis. Particularly, we elucidated the molecular mechanisms of STAT3-mediated gene regulation in angiogenic endothelial cells in response to external stimulations such as hypoxia and inflammation. The potential for STAT3 as a therapeutic target was also discussed. Overall, this review provides mechanistic insights for the roles of STAT3 signaling in tumor angiogenesis.

9.
Oncotarget ; 8(27): 44944-44959, 2017 Jul 04.
Article in English | MEDLINE | ID: mdl-28388546

ABSTRACT

Angiogenesis is crucial for tumor growth and metastasis. Cadmium (Cd) exposure is associated with elevated cancer risk and mortality. Such association is, at least in part, attributable to Cd-induced tumor angiogenesis. Nevertheless, the reported effects of Cd on tumor angiogenesis appear to be either stimulatory or inhibitory, depending on the concentrations. Ultra-low concentrations of Cd (<0.5 µM) inhibit endothelial nitric oxide synthase activation, leading to reduced endothelial nitric oxide production and attenuated tumor angiogenesis. In contrast, low-lose Cd (1-10 µM) up-regulates vascular endothelial growth factor (VEGF)-mediated tumor angiogenesis by exerting sub-apoptotic levels of oxidative stress on both tumor cells and endothelial cells (ECs). The consequent activation of protein kinase B/Akt, nuclear factor-κB, and mitogen-activated protein kinase signaling cascades mediate the increased secretion of VEGF by tumor cells and the up-regulated VEGF receptor-2 expression in ECs. Furthermore, Cd in high concentrations (>10 µM) induces EC apoptosis via the activation of caspase-3, resulting in destruction of tumor vasculature. In this review, we summarize the current knowledge concerning the roles of Cd in tumor angiogenesis, with a focus on molecular mechanisms underlying the dose dependent effects of Cd on various EC phenotypes.


Subject(s)
Cadmium/adverse effects , Environmental Pollutants/adverse effects , Neoplasms/etiology , Neoplasms/pathology , Neovascularization, Pathologic/chemically induced , Animals , Cadmium/administration & dosage , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Environmental Pollutants/administration & dosage , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
10.
Article in English | MEDLINE | ID: mdl-27739415

ABSTRACT

Cadmium (Cd) is a heavy metal and environmental pollutant. The kidney is the principal target organ of Cd exposure. Previously, we found that low concentration of Cd damages the integrity of the glomerular filtration barrier. However, little is known about the effects of Cd on renal mesangial cells, which provide structural support for the glomerular capillary loops and regulate intraglomerular blood flow. In this study, human renal mesangial cells (HRMCs) were cultured in the presence of serum and treated with 4 µM Cd. We found that Cd activates the c-Jun N-terminal kinase (JNK) pathway, and increases the protein levels of c-Jun and c-Fos. Cd treatment also induces a decrease in proliferation and an increase in apoptosis of HRMCs, but only the decrease in HRMC proliferation was reversed by pretreatment with SP600125, an inhibitor of the JNK pathway. In addition, Cd does not change the expression of α-smooth muscle actin and platelet-derived growth factor receptor-ß, the markers of mesangial cells, or the alignment of the filamentous actin (F-actin) cytoskeleton of HRMCs. Our data indicate that the JNK pathway mediates the inhibitory effects of Cd on HRMC proliferation.


Subject(s)
Cadmium/administration & dosage , Cadmium/pharmacology , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , JNK Mitogen-Activated Protein Kinases/drug effects , Mesangial Cells/drug effects , Animals , Apoptosis/drug effects , Cells, Cultured , Environmental Pollutants/pharmacology , Humans , MAP Kinase Signaling System
11.
Int J Mol Med ; 38(2): 417-22, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27315281

ABSTRACT

The kidney is one of the primary organs targeted by cadmium (Cd), a widely distributed environmental pollutant. The glomerular endothelium is the major component of the glomerular filtration barrier. However, the effects of Cd on glomerular endothelial cells remain largely unknown. For this purpose, we aimed to determine the effects of low dose Cd on the survival of human renal glomerular endothelial cells (HRGECs). Cultured HRGECs were exposed to 4 µM cadmium chloride (CdCl2) and examined at different time-points. We found that Cd activates the nuclear factor-κB (NF-κB) pathway without inducing the apoptosis of HRGECs. Pre-treating the cells with pyrrolidine dithiocarbamate (PDTC), a potent NF-κB inhibitor, prior to Cd exposure triggered extensive cell death (73.5%). In addition, Cd activates the c-Jun N-terminal kinase (JNK) pathway, and inhibition of the NF-κB pathway significantly elevates Cd-induced JNK phosphorylation in HRGECs (p<0.01). The combination treatment of PDTC and SP600125, a JNK pathway inhibitor, increased the survival of Cd-stimulated HRGECs compared with those cells treated with PDTC alone (p<0.05). Taken together, these findings demonstrate that the NF-κB pathway plays an essential role in maintaining the survival of Cd-exposed HRGECs.


Subject(s)
Cadmium/toxicity , Endothelial Cells/pathology , Kidney Glomerulus/pathology , NF-kappa B/metabolism , Signal Transduction/drug effects , Anthracenes/pharmacology , Apoptosis/drug effects , Cell Survival/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Pyrrolidines/pharmacology , Thiocarbamates/pharmacology
12.
J Appl Toxicol ; 36(2): 257-65, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26011702

ABSTRACT

The kidney is the principal organ targeted by exposure to cadmium (Cd), a well-known toxic metal. Even at a low level, Cd damages glomerular filtration. However, little is known about the effects of Cd on the glomerular endothelium, which performs the filtration function and directly interacts with Cd in blood plasma. In this study, we cultured human renal glomerular endothelial cells (HRGECs) in the presence of serum with treatment of a short term (1 h) and low concentration (1 µm) of Cd, which mimics the pattern of glomerular endothelium exposure to Cd in vivo. We found that this short-term, low-dose Cd exposure does not induce cytotoxicity, but increases permeability in HRGECs monolayers and redistributes adherens junction proteins vascular endothelial-cadherin and ß-catenin. Though short-term, low-dose Cd exposure activates all three major mitogen activated protein kinases, only the inhibitor of p38 mitogen activated protein kinase partially prevents Cd-induced hyperpermeability in HRGECs. Our data indicate that the presence of Cd in blood circulation might directly disrupt the glomerular endothelial cell barrier and contribute to the development of clinical symptoms of glomerular diseases.


Subject(s)
Cadmium/toxicity , Cells, Cultured/drug effects , Endothelial Cells/drug effects , Glomerular Filtration Rate/drug effects , Kidney/drug effects , Permeability/drug effects , Cadmium/blood , Cell Enlargement/drug effects , Cell Proliferation/drug effects , Humans
13.
Int J Environ Res Public Health ; 12(9): 10508-21, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26343694

ABSTRACT

Cadmium (Cd) is a heavy metal and environmental toxin. Exposure to Cd has been associated with a variety of human cancers. In this study, we performed in vitro assays to examine the effects of cadmium chloride (CdCl2) on A549 cells, a human lung adenocarcinoma cell line. Cd does not affect proliferation, migration, or apoptosis of A549 cells at concentrations of 0.1-10 µM. At 0.5 and 1 µM, Cd increases the expression of vascular endothelial growth factor (VEGF) (p < 0.05, p < 0.01, respectively), but not basic fibroblast growth factor (b-FGF) in A549 cells. The conditioned media were collected from the A549 cells treated with 1 µM Cd and were co-cultured with human umbilical vein endothelial cells (HUVECs). Upon treatment with the conditioned media, the proliferation and migration of HUVECs significantly increased (p < 0.01, p < 0.05, respectively), while apoptosis remained unchanged. In addition, 1 µM Cd increases the level of hypoxia inducible factor 1-α (HIF1-α), which is a positive regulator of VEGF expression. Although low-dose Cd does not directly affect the growth of lung adenocarcinoma cells, it might facilitate the development of tumors through its pro-angiogenic effects.


Subject(s)
Apoptosis/drug effects , Cadmium/toxicity , Cell Movement/drug effects , Cell Proliferation/drug effects , Environmental Pollutants/toxicity , Fibroblast Growth Factor 2/genetics , Vascular Endothelial Growth Factor A/genetics , Cell Line, Tumor , Fibroblast Growth Factor 2/metabolism , Humans , Vascular Endothelial Growth Factor A/metabolism
14.
Birth Defects Res A Clin Mol Teratol ; 103(12): 1039-45, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26408417

ABSTRACT

BACKGROUND: While it is known that cadmium-exposed embryonic cells have increased activation of c-Jun N-terminal kinase (JNK), the role of this stress signaling pathway in the embryotoxic response is not clear. Thus, the effects of modification of the transcription factor c-Jun, one of the downstream targets of JNK, on cadmium-induced embryotoxicity were investigated in primary cultures of mouse embryo limb bud cells. METHODS: Cultures of limb bud cells harvested on day 11 of gestation were pretreated with antisense oligonucleotides (ASO) to c-Jun to reduce its expression, and then incubated with cadmium in the form of cadmium chloride. Toxicity was measured through assessments of cell proliferation and differentiation, while the effectiveness of the ASO in reducing c-Jun was assessed through Western blotting using phosphorylation-specific antibodies. RESULTS: When cells were treated with ASO c-Jun, the total amounts of c-Jun and also cadmium-induced c-Jun activation were diminished. Cadmium-induced cytotoxicity, indicated by reduced cell numbers and differentiation, was found to decrease when cells were exposed to the antisense oligonucleotides to c-Jun. In addition, limb cell numbers and differentiation were also enhanced by exposure to ASO in the absence of cadmium. CONCLUSION: The JNK pathway, and particularly the downstream effector c-Jun, appears to play an important role in regulating cell survival and differentiation in mouse embryo limb bud cells both in the presence and absence of the toxic metal cadmium.


Subject(s)
Cadmium/toxicity , JNK Mitogen-Activated Protein Kinases/metabolism , Limb Buds/drug effects , Proto-Oncogene Proteins c-jun/metabolism , Animals , Cell Differentiation/drug effects , Cells, Cultured , Limb Buds/cytology , Limb Buds/enzymology , Limb Buds/metabolism , Mice , Phosphorylation
15.
Birth Defects Res A Clin Mol Teratol ; 88(9): 707-14, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20672348

ABSTRACT

INTRODUCTION: Cadmium (Cd(2+)) induces limb defects and other malformations in experimental animals. However, the mechanisms of the developmental toxicity of this metal are not fully understood. The ubiquitous intracellular tripeptide glutathione (GSH) protects nonembryonic cells from Cd(2+)-induced cell death and is essential for normal embryonic development. We predicted that pretreatment with GSH would prevent cytotoxicity in cultured mouse embryonic limb bud cells exposed to Cd(2+). Additionally, it was expected that GSH pretreatment would prevent the Cd(2+)-induced activation of the signaling molecule c-Jun N-terminal kinase (JNK), which becomes phosphorylated upon exposure to Cd(2+). METHODS: Primary micromass cultures of limb bud cells obtained from organogenesis-stage mouse embryos were treated with either Cd(2+) or GSH alone, or both Cd(2+) and GSH. Treatment with GSH commenced 4 hr prior to Cd(2+) treatment. RESULTS: Cell proliferation was inhibited by approximately 50% after exposure to 4 µM Cd(2+) for 5 days. Cd(2+) treatment also resulted in a dose-dependent increase in the intracellular GSH content as measured after 5 days of exposure. Pretreatment with 4 mM GSH for 4 hr prevented the Cd(2+)-induced inhibition of cell proliferation and differentiation and also inhibited a threefold activation of JNK induced by 4 µM Cd(2+) after 24 and 48 hr of exposure. CONCLUSION: Exogenous GSH protects cultured embryonic limb bud cells from Cd(2+)-induced inhibition of cell proliferation and differentiation, which is associated with the activation of JNK.


Subject(s)
Cadmium/toxicity , Glutathione/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Limb Buds/drug effects , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Chondrogenesis/drug effects , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Enzyme Activation/drug effects , Extracellular Matrix/drug effects , Glutathione/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , Limb Buds/cytology , Limb Buds/embryology , Limb Buds/metabolism , Limb Deformities, Congenital , Mice , Mice, Inbred BALB C , Oxidative Stress , Signal Transduction , Teratogens/toxicity
16.
Reprod Toxicol ; 29(3): 286-91, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20152897

ABSTRACT

While it is well accepted that mammalian embryos may develop abnormally in response to harmful environmental stresses, the molecular changes that embryonic cells undergo are not always clear. In the present study we examined the induction of mitogen-activated protein kinase (MAPK) pathways by cadmium chloride (CdCl(2)) in micromass cultures of limb bud cells from day 11 BALB/c mouse embryos. CdCl(2) exposure was found to cause a marked reduction in both cell survival and differentiation in these cultures. To study stress signalling pathways, cells were lysed and were examined for the phosphorylation of three MAPKs. It was shown that 4 microM CdCl(2) treatment of limb bud cells caused the phosphorylation of c-Jun N-terminal kinase (JNK), but not p38 or extracellular signal-regulated kinase (ERK), over a 120min period. The results indicate a specific activation of the JNK pathway in response to CdCl(2)-induced toxicity in mouse limb bud cells.


Subject(s)
Cadmium/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Animals , Cadmium Chloride/metabolism , Cadmium Chloride/toxicity , Cell Differentiation , Cell Survival/drug effects , Embryo, Mammalian/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Limb Buds/metabolism , Mice , Mice, Inbred BALB C , Phosphorylation , Signal Transduction
17.
Environ Toxicol Pharmacol ; 22(1): 1-7, 2006 Jul.
Article in English | MEDLINE | ID: mdl-21783678

ABSTRACT

Members of the c-Jun NH(2)-terminal kinase (JNK) signalling pathway have been found to be stimulated by a variety of stresses, including heavy metals, hyperthermia, and UV-irradiation. In the present study, we examined whether exposure of micromass cultures of mouse embryonic midbrain cells to a known teratogen, cadmium, leads to the phosphorylation and activation of JNK. Midbrain cells exposed to 0.5, 1, 2, or 4µM cadmium chloride (CdCl(2)) showed a dose-dependent decline in cell numbers, cell viability and differentiation after 5 days. In cells exposed to 4µM CdCl(2) for up to 1h, the level of phosphorylated JNK increased by 15min and peaked at 30min exposure time, as determined by a phospho-specific anti-JNK antibody, while the total amount of JNK protein did not change. This phosphorylated JNK was active, as shown by a corresponding increase in the level of c-Jun phosphorylated on Ser63 in a kinase assay. These results demonstrate that CdCl(2) induces a rapid and transient activation of the JNK pathway in primary embryonic neuron cell cultures.

SELECTION OF CITATIONS
SEARCH DETAIL
...