Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cells ; 12(5)2023 03 03.
Article in English | MEDLINE | ID: mdl-36899935

ABSTRACT

Neovascular age-related macular degeneration (AMD) is described as abnormal angiogenesis in the retina and the leaking of fluid and blood that generates a huge, dark, blind spot in the center of the visual field, causing severe vision loss in over 90% of patients. Bone marrow-derived endothelial progenitor cells (EPCs) contribute to pathologic angiogenesis. Gene expression profiles downloaded from the eyeIntegration v1.0 database for healthy retinas and retinas from patients with neovascular AMD identified significantly higher levels of EPC-specific markers (CD34, CD133) and blood vessel markers (CD31, VEGF) in the neovascular AMD retinas compared with healthy retinas. Melatonin is a hormone that is mainly secreted by the pineal gland, and is also produced in the retina. Whether melatonin affects vascular endothelial growth factor (VEGF)-induced EPC angiogenesis in neovascular AMD is unknown. Our study revealed that melatonin inhibits VEGF-induced stimulation of EPC migration and tube formation. By directly binding with the VEGFR2 extracellular domain, melatonin significantly and dose-dependently inhibited VEGF-induced PDGF-BB expression and angiogenesis in EPCs via c-Src and FAK, NF-κB and AP-1 signaling. The corneal alkali burn model demonstrated that melatonin markedly inhibited EPC angiogenesis and neovascular AMD. Melatonin appears promising for reducing EPC angiogenesis in neovascular AMD.


Subject(s)
Endothelial Progenitor Cells , Melatonin , Wet Macular Degeneration , Humans , Angiogenesis Inhibitors , Vascular Endothelial Growth Factor A , Visual Acuity
2.
Int J Biol Sci ; 19(3): 760-771, 2023.
Article in English | MEDLINE | ID: mdl-36778124

ABSTRACT

Background: Metastatic prostate cancer (PCa) predicts a poor prognosis and lower likelihood of survival. Osteoblasts (OBs) are known to be responsible for the synthesis and mineralization of bone, although it is unclear as to whether PCa in the prostate gland cooperates with OBs in bone to promote PCa malignant transformation. We aimed to elucidate how primary PCa cells cooperate with distal OBs and contribute to the vicious cycle that leads to metastatic PCa. Methods: N-cadherin, E-cadherin, and Twist protein expression were measured by Western blot. Twist translocation into the nucleus was detected by the immunofluorescence (IF) assay. Enzyme-linked immunosorbent assay (ELISA) detected protein levels in human serum samples. Levels of candidate protein expression were examined by the human cytokine array. Prostate tumor growth and metastasis were analyzed by orthotopic and metastatic prostate cancer models, respectively. Immunohistochemistry (IHC) staining was used to observe ADAM metallopeptidase domain 9 (ADAM9) and WNT1 inducible signaling pathway protein 1 (WISP-1) expression in tissue. Results: Our in vitro and in vivo analyses have now discovered that primary PCa expressing ADAM9 protein enables the transformation of OBs into PCa-associated osteoblasts (PCa-OBs), inducing WISP-1 secretion from PCa-OBs in the bone microenvironment. The upregulation of WISP-1 in bone provided feedback to primary PCa and promoted PCa cell aggressiveness via epithelial-mesenchymal transition (EMT) activity. Elevated levels of WISP-1 expression were detected in the serum of patients with PCa. ADAM9 levels were overexpressed in tumor tissue from PCa patients; ADAM9 blockade interrupted OB-induced release of WISP-1 and also suppressed primary tumor growth and distal metastasis in orthotopic PCa mouse models. Conclusion: Our study suggests that the ADAM9/WISP-1 axis assists with metastatic PCa progression. Thus, targeting the ADAM9/WISP-1 axis may help to prevent the malignant phenotypes of PCa cells.


Subject(s)
ADAM Proteins , Prostatic Neoplasms , Animals , Humans , Male , Mice , ADAM Proteins/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Membrane Proteins/genetics , Membrane Proteins/metabolism , Osteoblasts/metabolism , Prostatic Neoplasms/metabolism , Tumor Microenvironment , Up-Regulation
3.
J Pineal Res ; 72(3): e12793, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35174530

ABSTRACT

Advanced prostate cancer often develops into bone metastasis, which is characterized by aberrant bone formation with chronic pain and lower chances of survival. No treatment exists as yet for osteoblastic bone metastasis in prostate cancer. The indolamine melatonin (N-acetyl-5-methoxytryptamine) is a major regulator of the circadian rhythm. Melatonin has shown antiproliferative and antimetastatic activities but has not yet been shown to be active in osteoblastic bone lesions of prostate cancer. Our study investigations reveal that melatonin concentration-dependently decreases the migratory and invasive abilities of two osteoblastic prostate cancer cell lines by inhibiting FAK, c-Src, and NF-κB transcriptional activity via the melatonin MT1 receptor, which effectively inhibits integrin α2 ß1 expression. Melatonin therapy appears to offer therapeutic possibilities for reducing osteoblastic bone lesions in prostate cancer.


Subject(s)
Melatonin , Prostatic Neoplasms , Cell Line, Tumor , Humans , Integrin alpha2beta1/therapeutic use , Male , Melatonin/pharmacology , Melatonin/therapeutic use , NF-kappa B/metabolism , Prostatic Neoplasms/metabolism
4.
Oncogene ; 40(8): 1503-1515, 2021 02.
Article in English | MEDLINE | ID: mdl-33452455

ABSTRACT

Cancer-related bone erosion occurs frequently in bone metastasis and is associated with severe complications such as chronic bone pain, fractures, and lower survival rates. In recognition of the fact that the darkness hormone melatonin is capable of regulating bone homeostasis, we explored its therapeutic potential in bone metastasis. We found that melatonin directly reduces osteoclast differentiation, bone resorption activity and promotes apoptosis of mature osteoclasts. We also observed that melatonin inhibits RANKL production in lung and prostate cancer cells by downregulating the p38 MAPK pathway, which in turn prevents cancer-associated osteoclast differentiation. In lung and prostate bone metastasis models, twice-weekly melatonin treatment markedly reduced tumor volumes and numbers of osteolytic lesions. Melatonin also substantially lowered the numbers of TRAP-positive osteoclasts in tibia bone marrow and RANKL expression in tumor tissue. These findings show promise for melatonin in the treatment of bone metastases.


Subject(s)
Bone Neoplasms/drug therapy , Melatonin/pharmacology , Osteoclasts/drug effects , RANK Ligand/genetics , p38 Mitogen-Activated Protein Kinases/genetics , Animals , Bone Marrow Cells/drug effects , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Bone Resorption/drug therapy , Bone Resorption/genetics , Bone Resorption/pathology , Cell Differentiation/drug effects , Disease Models, Animal , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Macrophages/drug effects , Male , Mice , Osteoclasts/pathology , Osteolysis/drug therapy , Osteolysis/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RAW 264.7 Cells/drug effects , Signal Transduction/drug effects , Tartrate-Resistant Acid Phosphatase/genetics
5.
Environ Toxicol ; 36(3): 425-432, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33107671

ABSTRACT

Prostate cancer osteoblastic bone metastases are incurable and associated with chronic bone pain and a high mortality rate. Osteoclast-targeting drugs intended to prevent skeletal-related events associated with prostate cancer bone metastases do not prolong overall survival. Improved understanding of the bone-derived factors that contribute to prostate cancer osteoblastic bone metastases is required to design treatments that will improve morbidities and overall survival. Activated osteoblasts stimulate prostate cancer growth in bone. In this study, we report that prostate cancer conditioned medium (CM) promoted bone morphogenetic protein (BMP)-2, -4 and -7 production and the expression of osteogenic transcription factors Runx2 and osterix in osteoblasts. Treating the prostate cancer CM with antibody against CCN3 (nephroblastoma-overexpressed), a cysteine-rich protein that belongs to the CCN family, reduced all of these increases. Incubation of osteoblasts with CCN3 facilitated phosphorylation of GSK3ß and ß-catenin. GSK3ß and ß-catenin inhibitors or siRNAs all abolished CCN3-induced promotion of BMPs, Runx2 and osterix expression in osteoblasts. Our results indicate that prostate cancer-secreted CCN3 enhances BMP, Runx2 and osterix expression in osteoblasts via the GSK3ß and ß-catenin signaling pathways. This understanding of the role played by CCN3 in osteoblastic prostate bone metastasis may lead to more efficient targeted therapies.


Subject(s)
Glycogen Synthase Kinase 3 beta/metabolism , Nephroblastoma Overexpressed Protein/metabolism , Prostatic Neoplasms/metabolism , beta Catenin/metabolism , Animals , Bone Neoplasms/pathology , Cell Differentiation , Humans , Male , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteogenesis , Phosphorylation , Signal Transduction
6.
J Cell Physiol ; 236(5): 3979-3990, 2021 05.
Article in English | MEDLINE | ID: mdl-33251599

ABSTRACT

Prostate cancer has high metastatic potential. Men with higher urinary levels of the sleep hormone melatonin are much less likely to develop advanced prostate cancer compared with men with lower levels of melatonin. Melatonin has shown anticancer activity in experimental investigations. Nevertheless, the therapeutic effect of melatonin in metastatic prostate cancer has largely remained a mystery. Analyses of Gene Expression Omnibus data and human tissue samples indicated that levels of matrix metallopeptidase 13 (MMP-13) expression are higher in prostate cancer patients than in healthy cancer-free individuals. Mechanistic investigations revealed that melatonin inhibits MMP-13 expression and the migratory and invasive capacities of prostate cancer cells via the MT1 receptor and the phospholipase C, p38, and c-Jun signaling cascades. Importantly, tumor growth rate and metastasis to distant organs were suppressed by melatonin in an orthotopic prostate cancer model. This is the first demonstration showing that melatonin impedes metastasis of prostate cancer by suppressing MMP-13 expression in both in vitro and in vivo models. Thus, melatonin is promising in the management of prostate cancer metastasis and deserves to undergo clinical investigations.


Subject(s)
Matrix Metalloproteinase 13/metabolism , Melatonin/pharmacology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Animals , Cell Death/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Down-Regulation/drug effects , Humans , Male , Mice, SCID , Models, Biological , Neoplasm Invasiveness , Neoplasm Metastasis , Proto-Oncogene Proteins c-jun/metabolism , Receptors, Melatonin/metabolism , Type C Phospholipases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Biochem Pharmacol ; 155: 537-546, 2018 09.
Article in English | MEDLINE | ID: mdl-30031810

ABSTRACT

Thrombospondin (TSP)-2, a matricellular glycoprotein of the TSP family, regulates multiple biological functions, including proliferation, angiogenesis, cell adhesion, and extracellular matrix (ECM) modeling. The clinical relevance of TSP-2 has been explored in many different cancers. TSP-2 expression levels vary between different cancer types, and their role in tumor progression remains controversial. Although previous studies have reported higher serum TSP-2 levels in patients with non-small cell lung cancer, the role of TSP-2 in lung cancer progression remains to be addressed. A total of 585 lung adenocarcinoma datasets, including mRNA sequencing and clinical data, were retrieved from The Cancer Genome Atlas (TCGA). Forty paired adjacent normal tissues and lung tumor tissue datasets were used to examine TSP-2 expression levels. Tumor microarray were performed with immunohistochemical staining to examine TSP-2 expression in lung cancer patients. Transwell migration assay, quantitative real-time PCR and Western blot were used to investigate molecular mechanism of TSP-2 in lung cancer cell. TSP-2 promotes matrix metalloproteinase-13 (MMP-13) expression, cell migration, and cell invasion by mediating integrin αvß3/FAK/Akt/NF-κB signal transduction. Using TSP-2 knockdown stable cell lines, we found that TSP-2 knockdown reduces MMP-13 expression and cell mobility. When we manipulated the tumor tissue microarray and TCGA datasets to investigate the clinical relevance of TSP-2, we found high TSP-2 expression levels in lung cancer specimens. The present study demonstrates that TSP-2 regulates cell mobility through MMP-13 expression in lung cancer cells. In addition, TSP-2 expression was associated with MMP-13 expression and poor prognosis in lung cancer. TSP-2 may therefore be a promising novel target for lung cancer treatment.


Subject(s)
Lung Neoplasms/chemically induced , Lung Neoplasms/enzymology , Matrix Metalloproteinase 13/biosynthesis , Thrombospondins/biosynthesis , A549 Cells , Cell Movement/drug effects , Cell Movement/physiology , Enzyme Induction/drug effects , Enzyme Induction/physiology , Humans , Lung Neoplasms/pathology , Neoplasm Invasiveness/pathology , Thrombospondins/toxicity
8.
J Hematol Oncol ; 10(1): 33, 2017 01 25.
Article in English | MEDLINE | ID: mdl-28122633

ABSTRACT

BACKGROUND: Thrombospondin-2 (TSP-2) is a secreted matricellular glycoprotein that is found to mediate cell-to-extracellular matrix attachment and participates in many physiological and pathological processes. The expression profile of TSP-2 on tumors is controversial, and it up-regulates in some cancers, whereas it down-regulates in others, suggesting that the functional role of TSP-2 on tumors is still uncertain. METHODS: The expression of TSP-2 on prostate cancer progression was determined in the tissue array by the immunohistochemistry. The molecular mechanism of TSP-2 on prostate cancer (PCa) metastasis was investigated through pharmaceutical inhibitors, siRNAs, and miRNAs analyses. The role of TSP-2 on PCa metastasis in vivo was verified through xenograft in vivo imaging system. RESULTS: Based on the gene expression omnibus database and immunohistochemistry, we found that TSP-2 increased with the progression of PCa, especially in metastatic PCa and is correlated with the matrix metalloproteinase-2 (MMP-2) expression. Additionally, through binding to CD36 and integrin ανß3, TSP-2 increased cell migration and MMP-2 expression. With inhibition of p38, ERK, and JNK, the TSP-2-induced cell migration and MMP-2 expression were abolished, indicating that the TSP-2's effect on PCa is MAPK dependent. Moreover, the microRNA-376c (miR-376c) was significantly decreased by the TSP-2 treatment. Furthermore, the TSP-2-induced MMP-2 expression and the subsequent cell motility were suppressed upon miR-376c mimic stimulation. On the other hand, the animal studies revealed that the bone metastasis was abolished when TSP-2 was stably knocked down in PCa cells. CONCLUSIONS: Taken together, our results indicate that TSP-2 enhances the migration of PCa cells by increasing MMP-2 expression through down-regulation of miR-376c expression. Therefore, TSP-2 may represent a promising new target for treating PCa.


Subject(s)
Bone Neoplasms/secondary , Matrix Metalloproteinase 2/genetics , MicroRNAs/genetics , Prostatic Neoplasms/pathology , Thrombospondins/physiology , Cell Line , Cell Movement , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...