Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Pharmaceutics ; 15(7)2023 Jul 15.
Article in English | MEDLINE | ID: mdl-37514146

ABSTRACT

OBJECTIVES: Pigment epithelium-derived factor (PEDF) has been recently linked to insulin resistance and is capable of differentiating myocytes to bone. We examined in more detail the intricate signalling of the insulin pathway influenced by PEDF in skeletal myocytes. We tested whether this serpin is also capable of generating de novo bone from adipocytes in vitro and in vivo, and how the anticancer drug doxorubicin links with PEDF and cellular metabolism. METHODS AND KEY FINDINGS: We demonstrate that PEDF can inhibit phosphorylation of insulin receptor (IR) and insulin receptor substrate (IRS) in skeletal myocytes. PEDF constitutively activates p42/44 MAPK/Erk, but paradoxically does not affect mitogenic signalling. PEDF did not perturb either mitochondrial activity or proliferation in cells representing mesenchymal stem cells, cardiomyocytes, and skeletal myocytes and adipocytes. PEDF induced transdifferentiation of adipocytes to osteoblasts, promoting bone formation in cultured adipocytes in vitro and gelfoam fatpad implants in vivo. Bone formation in white adipose tissue (WAT) was better than in brown adipose tissue (BAT). The frontline anticancer drug doxorubicin increased levels of PEDF in a human breast cancer cell line, mirroring the in vivo finding where cardiac muscle tissue was stained increasingly for PEDF as the dose of doxorubicin increased in mice. PEDF also increased levels of reactive oxygen species (ROS) and glutathione (GSH) in the breast cancer cell line. CONCLUSIONS: PEDF may be used to regenerate bone from adipose tissue in cases of trauma such as fractures or bone cancers. The increased presence of PEDF in doxorubicin-treated tumour cells need further exploration, and could be useful therapeutically in future. The safety of PEDF administration in vivo was further demonstrated in this study.

2.
J Inflamm Res ; 14: 1019-1028, 2021.
Article in English | MEDLINE | ID: mdl-33790617

ABSTRACT

INTRODUCTION: Inflammation and bone erosion are processes key to the pathogenesis of rheumatoid arthritis, a systemic autoimmune disease causing progressive disability and pain, impacting around 1.3 million people in the United States alone. However, many patients do not respond sufficiently to existing therapies or benefit is not sustained and alternate therapeutic approaches are lacking. We recently identified the dibenzoxazepinone BT2, which inhibits ERK phosphorylation, from a high-throughput chemical screen and identified its ability to inhibit angiogenesis and vascular leakiness. METHODS: Here we evaluated BT2 for potential anti-inflammatory activity in in vitro models of human monocytic-endothelial cell adhesion, monocytic cell extravasation and collagen antibody-induced arthritis in mice. RESULTS: BT2 inhibits human monocytic cell adhesion to IL-1ß-treated human endothelial cells and inhibits monocytic transendothelial migration toward MCP-1. In mice rendered arthritic, single systemic administration of BT2 prevented footpad swelling, bone destruction and TRAP+ cells in the joints. BT2 suppressed inducible circulating levels of IL-1ß, IL-2 and IL-6 to normal levels without affecting levels of IL-4 or IL-10 among other cytokines. BT2 also inhibited the expression of pro-inflammatory adhesion molecules ICAM-1 and VCAM-1 in arthritic joints. There was no evidence of toxicity following intraperitoneal, gavage or intraarticular administration of BT2. CONCLUSION: BT2 is a novel small molecule inhibitor of joint inflammation, bone erosion, pro-inflammatory cytokine and adhesion molecule expression. This suggests the potential clinical utility of BT2 as a new anti-inflammatory agent.

3.
Sci Adv ; 6(31): eaaz7815, 2020 07.
Article in English | MEDLINE | ID: mdl-32923607

ABSTRACT

Vascular permeability and angiogenesis underpin neovascular age-related macular degeneration and diabetic retinopathy. While anti-VEGF therapies are widely used clinically, many patients do not respond optimally, or at all, and small-molecule therapies are lacking. Here, we identified a dibenzoxazepinone BT2 that inhibits endothelial cell proliferation, migration, wound repair in vitro, network formation, and angiogenesis in mice bearing Matrigel plugs. BT2 interacts with MEK1 and inhibits ERK phosphorylation and the expression of FosB/ΔFosB, VCAM-1, and many genes involved in proliferation, migration, angiogenesis, and inflammation. BT2 reduced retinal vascular leakage following rat choroidal laser trauma and rabbit intravitreal VEGF-A165 administration. BT2 suppressed retinal CD31, pERK, VCAM-1, and VEGF-A165 expression. BT2 reduced retinal leakage in rats at least as effectively as aflibercept, a first-line therapy for nAMD/DR. BT2 withstands boiling or autoclaving and several months' storage at 22°C. BT2 is a new small-molecule inhibitor of vascular permeability and angiogenesis.


Subject(s)
Capillary Permeability , Vascular Cell Adhesion Molecule-1 , Angiogenesis Inhibitors/pharmacology , Animals , Humans , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Proto-Oncogene Proteins c-fos/metabolism , Rabbits , Rats , Vascular Cell Adhesion Molecule-1/metabolism , Vascular Cell Adhesion Molecule-1/pharmacology , Vascular Endothelial Growth Factor A/metabolism
4.
Cancer Lett ; 459: 268-276, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31128215

ABSTRACT

The incidence of melanoma is increasing faster than any other cancer. In recent years, treatment of melanoma and a range of other deadly cancers has involved immunotherapy with programmed cell death protein-1 (PD-1)/PD-1 ligand (PD-L1) checkpoint blockade which has improved survival. However, many patients do not respond or have partial response, survival benefit is in the order of months and all available PD-1/PD-L1 strategies are antibodies requiring intravenous infusion. There are no clinically approved small molecule pharmacologic inhibitors of the PD-1/PD-L1 system. The benzimidazole derivative flubendazole is a widely used anthelmintic available over the counter in Europe. Here we demonstrate the ability of flubendazole to inhibit human melanoma growth and spread in mice. Flubendazole's ability to block tumor growth and spread was comparable to paclitaxel. Anti-tumor effects were observed when flubendazole was delivered systemically not locally. Flubendazole inhibited CD31/PECAM-1 staining indicating suppression of tumor angiogenesis. Most surprisingly, flubendazole inhibited PD-1 levels within the tumors, but not PD-L1. Western blotting and flow cytometry revealed that flubendazole inhibits PD-1 expression in cultured melanoma cells. Flubendazole also reduced myeloid-derived suppressor cell (MDSC) levels in tumor tissue. Further we found that flubendazole inhibited active (phospho-Tyr705) signal transducer and activator of transcription (STAT3), an upstream regulator of PD-1 expression. These findings uncover that flubendazole is a novel small molecule inhibitor of not only melanoma growth and spread but also of PD-1 and MDSC.


Subject(s)
Mebendazole/analogs & derivatives , Melanoma/drug therapy , Myeloid-Derived Suppressor Cells/drug effects , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Antinematodal Agents/pharmacology , Antineoplastic Agents/pharmacology , Cell Growth Processes/drug effects , Cell Line, Tumor , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Mebendazole/pharmacology , Melanoma/blood supply , Melanoma/pathology , Mice , Mice, SCID , Myeloid-Derived Suppressor Cells/pathology , Neoplasm Metastasis , Neovascularization, Pathologic/drug therapy , Random Allocation , STAT3 Transcription Factor/metabolism , Xenograft Model Antitumor Assays
5.
Bone ; 124: 103-117, 2019 07.
Article in English | MEDLINE | ID: mdl-31028961

ABSTRACT

Pigment epithelium-derived factor (PEDF) is known for its osteogenic properties, but its effects against primary and secondary bone tumors have not comprehensively been demonstrated. We show the ubiquitous expression of PEDF in murine embryonic tissue. Continuous administration of PEDF in pregnant mice for five days did not adversely affect foetal health, despite PEDF's known potent antiangiogenic properties. In the case of the devastating childhood bone cancer osteosarcoma, PEDF has direct anticancer activity per se, and protects against the toxicity of doxorubicin in the heart, small intestine and testes. PEDF demonstrated anti-proliferative and pro-apoptotic effects against human prostate and breast cancer cells, tumors which are known to metastasize to bone as the preferred secondary site. Caspase-2 was activated in both tumor cell types by PEDF. In models of prostate and breast cancer in bone, PEDF significantly reduced tumor volumes. When combined with zoledronic acid, continuously-administered PEDF significantly reduced breast tumor volume at the bone, and was able to preserve the quality of bone better than the combination therapy. These multiple positive findings make PEDF an ideal endogenous and safe biological for possible future clinical testing.


Subject(s)
Bone Neoplasms/drug therapy , Doxorubicin/adverse effects , Doxorubicin/therapeutic use , Eye Proteins/therapeutic use , Nerve Growth Factors/therapeutic use , Serpins/therapeutic use , Animals , Apoptosis/drug effects , Bone Neoplasms/pathology , Cardiotonic Agents , Caspase 2/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Doxorubicin/pharmacology , Enzyme Activation/drug effects , Eye Proteins/pharmacology , Fetus/drug effects , Fetus/metabolism , Humans , Mice, Inbred BALB C , Nerve Growth Factors/pharmacology , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Serpins/pharmacology , Zoledronic Acid/pharmacology
6.
Curr Drug Targets ; 19(5): 467-478, 2018.
Article in English | MEDLINE | ID: mdl-26424392

ABSTRACT

BACKGROUND: Bone defects can be severely debilitating and reduce quality of life. Osteoregeneration can alleviate some of the complications in bony defects. For therapeutic use in future, a single factor that can cause potent bone regeneration is highly preferred as it will be more costeffective, any off-target effects will be more easily monitored and potentially managed, and for ease of administration which would lead to better patient compliance and satisfaction. OBJECTIVE: We demonstrate that pigment epithelium-derived factor (PEDF), one such factor that is known to be potent against angiogenesis, promotes osteoblastogenesis in mesenchymal stem cells in vitro, but does not need co-encapsulation of cells in alginate bead scaffolds for osteogeneration in vivo. RESULTS: Osteogenic differentiation by PEDF in vitro was confirmed with immunoblotting and immunocytochemical staining for bone markers (alkaline phosphatase, osteocalcin, osteopontin, collagen I), calcified mineral deposition, and assay for alkaline phosphatase activity. PEDF-mediated bone formation in a muscle pocket in vivo model was confirmed by microcomputed tomography (microCT), histology (haematoxylin and eosin, Alcian blue staining), immunostaining for bone markers and for collagen I-processing proteins (heat shock protein 47 and membrane type I matrix metalloproteinase). CONCLUSION: PEDF therefore presents itself as a promising biological for osteogeneration.


Subject(s)
Alginates/chemistry , Bone and Bones/metabolism , Eye Proteins/administration & dosage , Mesenchymal Stem Cells/cytology , Nerve Growth Factors/administration & dosage , Serpins/administration & dosage , Animals , Biomarkers/metabolism , Bone and Bones/diagnostic imaging , Bone and Bones/drug effects , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Survival , Cells, Cultured , Disease Models, Animal , Drug Compounding , Eye Proteins/chemistry , Eye Proteins/pharmacology , Humans , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Mice , Nerve Growth Factors/chemistry , Nerve Growth Factors/pharmacology , Osteogenesis/drug effects , Serpins/chemistry , Serpins/pharmacology , X-Ray Microtomography
7.
Mol Cell Endocrinol ; 472: 159-166, 2018 09 05.
Article in English | MEDLINE | ID: mdl-29258756

ABSTRACT

Extensive bone defects arising as a result of trauma, infection and tumour resection and other bone pathologies necessitates the identification of effective strategies in the form of tissue engineering, gene therapy and osteoinductive agents to enhance the bone repair process. PEDF is a multifunctional glycoprotein which plays an important role in regulating osteoblastic differentiation and bone formation. PEDF treatment of mice and human skeletal myocytes at physiological concentration inhibited myogenic differentiation and activated Erk1/2 MAPK- dependent osteogenic transdifferentiation of myocytes. In mice, insulin, a promoter of bone regeneration, attenuated PEDF-induced expression of osteogenic markers such as osteocalcin, alkaline phosphatase and mineralisation for bone formation in the muscle and surrounding adipose tissue. These results provide new insights into the molecular aspects of the antagonising effect of insulin on PEDF-dependent modulation of the differentiation commitment of musculoskeletal environment into osteogenesis, and suggest that PEDF may be developed as an effective clinical therapy for bone regeneration as its heterotopic ossification can be controlled via co-administration of insulin.


Subject(s)
Cell Lineage/drug effects , Eye Proteins/pharmacology , Insulin/pharmacology , Muscle Cells/pathology , Nerve Growth Factors/pharmacology , Ossification, Heterotopic/pathology , Serpins/pharmacology , Animals , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Humans , MAP Kinase Signaling System/drug effects , Mice, Inbred BALB C , Models, Biological , Muscle Cells/drug effects , Muscle Cells/metabolism , MyoD Protein/metabolism , Myogenin/metabolism , Osteogenesis/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism
8.
Crit Rev Clin Lab Sci ; 53(3): 166-83, 2016.
Article in English | MEDLINE | ID: mdl-26678521

ABSTRACT

An increasing body of evidence suggests that cerebrovascular dysfunction and microvessel disease precede the evolution of hallmark pathological features that characterise Alzheimer's disease (AD), consistent with a causal association for onset or progression. Recent studies, principally in genetically unmanipulated animal models, suggest that chronic ingestion of diets enriched in saturated fats and cholesterol may compromise blood-brain barrier (BBB) integrity resulting in inappropriate blood-to-brain extravasation of plasma proteins, including lipid macromolecules that may be enriched in amyloid-ß (Aß). Brain parenchymal retention of blood proteins and lipoprotein bound Aß is associated with heightened neurovascular inflammation, altered redox homeostasis and nitric oxide (NO) metabolism. Therefore, it is a reasonable proposition that lipid-lowering agents may positively modulate BBB integrity and by extension attenuate risk or progression of AD. In addition to their robust lipid lowering properties, reported beneficial effects of lipid-lowering agents were attributed to their pleiotropic properties via modulation of inflammation, oxidative stress, NO and Aß metabolism. The review is a contemporary consideration of a complex body of literature intended to synthesise focussed consideration of mechanisms central to regulation of BBB function and integrity. Emphasis is given to dietary fat driven significant epidemiological evidence consistent with heightened risk amongst populations consuming greater amounts of saturated fats and cholesterol. In addition, potential neurovascular benefits associated with the use of hypolipidemic statins, probucol and fenofibrate are also presented in the context of lipid-lowering and pleiotropic properties.


Subject(s)
Alzheimer Disease/physiopathology , Brain/blood supply , Brain/physiopathology , Capillaries/physiopathology , Dietary Fats/adverse effects , Risk Reduction Behavior , Alzheimer Disease/drug therapy , Animals , Anti-Inflammatory Agents/therapeutic use , Humans
9.
Nicotine Tob Res ; 17(12): 1436-41, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25744960

ABSTRACT

INTRODUCTION: Emerging evidence suggests that integrity of blood-brain barrier (BBB) is pivotal to pathology and pathogenesis of vascular-based neurodegenerative disorders. We have recently reported BBB protective effects of nutraceutical agents with anti-inflammatory properties in an established dietary-induced BBB dysfunction model. Studies also reported that nicotine exhibits anti-oxidative/-inflammatory effects and improve cognitive impairment in Alzheimer's disease. However there has been no studies reporting the effect of nicotine on high-fat-induced BBB dysfunction. METHODS: In the present study, we investigated the effect of nicotine on BBB integrity and neuro-inflammation in an established mouse model of BBB disruption induced by a diet enriched in saturated fatty acids (SFA). RESULTS: Wild-type C57BL/6J mice were fed chow enriched in SFA (23% w/w) with/without nicotine for 10 weeks. Compared to mice maintained on SFA-free and low-fat (LF) chow (4% w/w), capillary permeability indicated by the parenchymal extravasation of plasma derived IgG, was significantly greater in the SFA treatment group. Nicotine provided concomitantly with the SFA diet significantly attenuated IgG extravasation, however it remained significantly greater than LF-controls. Markers of neurovascular inflammation glial fibrillary acidic protein, cyclooxygenase-2, and glucose regulated protein 78 remained exaggerated in SFA+nicotine treated mice compared to LF-controls. Nicotine did however modestly, but not significantly, improve plasma total anti-oxidative status in SFA fed mice. CONCLUSION: Nicotine moderately attenuated BBB disruption induced by chronic ingestion of high-SFA diet, but had no significant effect on neuroinflammation per se.


Subject(s)
Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Dietary Fats/toxicity , Fatty Acids/toxicity , Nicotine/administration & dosage , Animals , Blood-Brain Barrier/metabolism , Dietary Fats/administration & dosage , Fatty Acids/administration & dosage , Female , Mice , Mice, Inbred C57BL
10.
Immun Ageing ; 12: 2, 2015.
Article in English | MEDLINE | ID: mdl-25784952

ABSTRACT

BACKGROUND: Functional loss of blood-brain barrier (BBB) is suggested to be pivotal to pathogenesis and pathology of vascular-based neurodegenerative disorders such as Alzheimer's disease. We recently reported in wild-type mice maintained on standard diets, progressive deterioration of capillary function with aging concomitant with heightened neuroinflammation. However, the mice used in this study were relatively young (12 months of age) and potential mechanisms for loss of capillary integrity were not investigated per se. The current study therefore extended the previous finding to investigate the effect of aging on BBB integrity in aged mice at 24 months and its potential underlying molecular mechanisms. RESULTS: Immunomicroscopy analyses confirmed significantly increased capillary permeability with heightened neuroinflammation in naturally aged 24-month old mice compared to young control at 3 months of age. Aged mice showed significant attenuation in the expression of BBB tight junction proteins, occludin-1 and to lesser extent ZO-1 compared to young mice. In addition, TNF-α in cerebral endothelial cells of aged mice was significantly elevated compared to controls and this was associated with heightened peripheral inflammation. The expression of ICAM-1 and VCAM-1 remained unelevated, and no sign of leukocyte recruitment was observed in aged mice. CONCLUSION: The BBB breakdown that occurs during ordinary aging is associated with inflammation and disruption of tight junction complex assembly but not through leukocyte trafficking.

11.
J Pharm Pharmacol ; 66(11): 1586-92, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25175851

ABSTRACT

OBJECTIVE: Pigment epithelium-derived factor (PEDF) has proven anti-osteosarcoma activity. However, the mechanism(s) underpinning its ability to reduce primary bone tumour (osteosarcoma) metastasis is unknown. METHODS: Adult and fetal murine bone were immunostained for PEDF, collagen I (major protein in bone) and its processing proteins, heat shock protein 47 (HSP47, a chaperone protein for collagen I), membrane type I matrix metalloproteinase (MT1-MMP, a collagenase), and matrix metalloproteinase 2 (MMP-2, which is activated by MT1-MMP). Immunoblotting and immunocytochemistry were used to observe levels of the above biomarkers when human osteosarcoma cells were treated with PEDF. KEY FINDINGS: Immunohistochemical staining in adult and fetal bone mirrors collagen I. PEDF localised to ridges of trabecular bone in tibial cortex and to megakaryocytes within bone marrow. Second, we observed that PEDF upregulates collagen I, HSP47 and MT1-MMP, while downregulating MMP-2 in osteosarcoma cells in vitro. CONCLUSION: PEDF is a promising antagonist to osteosarcoma cell metastasis via downregulation of MMP-2, and can induce tumour cells to further adopt differentiative properties, thereby possibly reducing their aggressive growth in vitro and in vivo.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Neoplasms/metabolism , Collagen Type I/metabolism , Eye Proteins/pharmacology , HSP47 Heat-Shock Proteins/metabolism , Matrix Metalloproteinase 2/metabolism , Nerve Growth Factors/pharmacology , Osteosarcoma/metabolism , Serpins/pharmacology , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Biomarkers/metabolism , Bone Marrow/metabolism , Bone Neoplasms/drug therapy , Cell Culture Techniques , Cell Line, Tumor , Down-Regulation , Eye Proteins/metabolism , Eye Proteins/therapeutic use , Fetus , Humans , Matrix Metalloproteinase 1/metabolism , Mice, Inbred BALB C , Nerve Growth Factors/metabolism , Nerve Growth Factors/therapeutic use , Osteosarcoma/drug therapy , Serpins/metabolism , Serpins/therapeutic use , Tibia/drug effects , Tibia/metabolism
12.
Biomaterials ; 35(27): 7828-38, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24947230

ABSTRACT

Bone defects caused by fractures or cancer-mediated destruction are debilitating. Chitosan is commonly used in scaffold matrices for bone healing, but rarely as a free drug. We demonstrate that free chitosan promotes osteoblast proliferation and osteogenesis in mesenchymal stem cells, increases osteopontin and collagen I expression, and reduces osteoclastogenesis. Chitosan inhibits invasion of endothelial cells, downregulating uPA/R, MT1-MMP, cdc42 and Rac1. Better healing of bone fractures with greater trabecular bone formation was observed in mice treated with chitosan. Chitosan induces apoptosis in osteotropic prostate and breast cancer cells via caspase-2 and -3 activation, and reduces their establishment in bone. Chitosan is pro-apoptotic in osteosarcoma cells, but not their normal counterpart, osteoblasts, or chondrosarcoma cells. Systemic delivery of chitosan does not perturb angiogenesis, bone volume or instinctive behaviour in pregnant mice, but decreases foetal length and changes pancreatic secretory acini. With certain controls in place, chitosan could be useful for bone trauma management.


Subject(s)
Bone Neoplasms/drug therapy , Bone and Bones/pathology , Chitosan/therapeutic use , Wounds and Injuries/drug therapy , Animals , Apoptosis/drug effects , Bone Neoplasms/enzymology , Bone Neoplasms/pathology , Bone and Bones/drug effects , Caspase 2/metabolism , Cell Line, Tumor , Chitosan/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/pathology , Female , Fetus/drug effects , Fracture Healing/drug effects , Fractures, Bone/drug therapy , Fractures, Bone/pathology , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/pathology , Mice, Inbred BALB C , Osteoblasts/drug effects , Osteoblasts/pathology , Osteoclasts/drug effects , Osteoclasts/pathology , Pregnancy , Rats
13.
J Endocrinol ; 222(3): R129-39, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24928938

ABSTRACT

Pigment epithelium-derived factor (PEDF) is a pluripotent glycoprotein belonging to the serpin family. PEDF can stimulate several physiological processes such as angiogenesis, cell proliferation, and survival. Oxidative stress plays an important role in the occurrence of diabetic retinopathy (DR), which is the major cause of blindness in young diabetic adults. PEDF plays a protective role in DR and there is accumulating evidence of the neuroprotective effect of PEDF. In this paper, we review the role of PEDF and the mechanisms involved in its antioxidative, anti-inflammatory, and neuroprotective properties.


Subject(s)
Diabetic Neuropathies/metabolism , Diabetic Neuropathies/prevention & control , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/prevention & control , Eye Proteins/metabolism , Nerve Growth Factors/metabolism , Reactive Oxygen Species/metabolism , Serpins/metabolism , Antioxidants/metabolism , Apoptosis , Diabetic Neuropathies/etiology , Diabetic Retinopathy/etiology , Endothelial Cells/metabolism , Humans , Inflammation/etiology , Inflammation/metabolism , Inflammation/prevention & control , Models, Biological , NADP/metabolism , Neuroprotective Agents/metabolism , Oxidative Stress , Pericytes/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , bcl-2-Associated X Protein/metabolism
14.
J Biomed Biotechnol ; 2012: 239091, 2012.
Article in English | MEDLINE | ID: mdl-22675247

ABSTRACT

Encoded by a single gene, PEDF is a 50 kDa glycoprotein that is highly conserved and is widely expressed among many tissues. Most secreted PEDF deposits within the extracellular matrix, with cell-type-specific functions. While traditionally PEDF is known as a strong antiangiogenic factor, more recently, as this paper highlights, PEDF has been linked with stem cell biology, and there is now accumulating evidence demonstrating the effects of PEDF in a variety of stem cells, mainly in supporting stem cell survival and maintaining multipotency.


Subject(s)
Eye Proteins/physiology , Nerve Growth Factors/physiology , Serpins/physiology , Stem Cells/physiology , Animals , Cell Biology , Humans , Stem Cell Research
15.
Chem Biol Drug Des ; 78(6): 909-12, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21722318

ABSTRACT

DNAzymes (DNA enzymes and deoxyribozymes) are synthetic, single-stranded DNA-based catalysts engineered to bind to their complementary sequence in a target messenger RNA (mRNA) through Watson-Crick rules for base-pairing and cleave the mRNA at predetermined phosphodiester linkages. Dz13, a DNAzyme that cleaves c-Jun mRNA, has been found to have efficacious effects against tumours directly, activity against tumour-induced angiogenesis, inhibition of neointima formation after arterial injury and control of inflammatory responses. Recent studies in endothelial cells demonstrate that the off-target effects of Dz13 may in fact be driving some of these potentially therapeutic effects, although no mechanisms have been clearly defined in tumour cells. Recent data show that Dz13 is capable of inhibiting more types of tumours and potently induces apoptosis in a panel of tumour cell lines. Hand-in-hand with in vivo testing, Dz13 has been formulated into a biocompatible nanoparticle, enabling its full potential to be realized. Its chemistry is partly responsible for its activity against tumour cells, but it is safe to use in vivo and surprisingly shows little harmful effects against normal cells. These findings provide hope that Dz13 may be useful clinically for the treatment of a variety of cancers.


Subject(s)
DNA, Catalytic/genetics , Down-Regulation , Neoplasms/metabolism , Proto-Oncogene Proteins c-jun/genetics , Animals , Apoptosis , Cell Death , DNA, Catalytic/metabolism , Humans , Neoplasms/genetics , Neoplasms/pathology , Neovascularization, Pathologic , Proto-Oncogene Proteins c-jun/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...