Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Extracell Vesicle ; 12022 Dec.
Article in English | MEDLINE | ID: mdl-37503329

ABSTRACT

Dysregulated Myc signaling is a key oncogenic pathway in glioblastoma multiforme (GBM). Yet, effective therapeutic targeting of Myc continues to be challenging. Here, we demonstrate that exosomes generated from human bone marrow mesenchymal stem cells (MSCs) engineered to encapsulate siRNAs targeting Myc (iExo-Myc) localize to orthotopic GBM tumors in mice. Treatment of late stage GBM tumors with iExo-Myc inhibits proliferation and angiogenesis, suppresses tumor growth, and extends survival. Transcriptional profiling of tumors reveals that the mesenchymal transition and estrogen receptor signaling pathways are impacted by Myc inhibition. Single nuclei RNA sequencing (snRNA-seq) shows that iExo-Myc treatment induces transcriptional repression of multiple growth factor and interleukin signaling pathways, triggering a mesenchymal to proneural transition and shifting the cellular landscape of the tumor. These data confirm that Myc is an effective anti-glioma target and that iExo-Myc offers a feasible, readily translational strategy to inhibit challenging oncogene targets for the treatment of brain tumors.

2.
Mol Ther Methods Clin Dev ; 22: 360-376, 2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34514028

ABSTRACT

Bladder cancer (BC), a heterogeneous disease characterized by high recurrence rates, is diagnosed and monitored by cystoscopy. Accurate clinical staging based on biopsy remains a challenge, and additional, objective diagnostic tools are needed urgently. We used exosomal DNA (exoDNA) as an analyte to examine cancer-associated mutations and compared the diagnostic utility of exoDNA from urine and serum of individuals with BC. In contrast to urine exosomes from healthy individuals, urine exosomes from individuals with BC contained significant amounts of DNA. Whole-exome sequencing of DNA from matched urine and serum exosomes, bladder tumors, and normal tissue (peripheral blood mononuclear cells) identified exonic and 3' UTR variants in frequently mutated genes in BC, detectable in urine exoDNA and matched tumor samples. Further analyses identified somatic variants in driver genes, unique to urine exoDNA, possibly because of the inherent intra-tumoral heterogeneity of BC, which is not fully represented in random small biopsies. Multiple variants were also found in untranslated portions of the genome, such as microRNA (miRNA)-binding regions of the KRAS gene. Gene network analyses revealed that exoDNA is associated with cancer, inflammation, and immunity in BC exosomes. Our findings show utility of exoDNA as an objective, non-invasive strategy to identify novel biomarkers and targets for BC.

3.
Exp Eye Res ; 188: 107798, 2019 11.
Article in English | MEDLINE | ID: mdl-31520600

ABSTRACT

Abnormal migration and proliferation of endothelial cells (EC) drive neovascular retinopathies. While anti-VEGF treatment slows progression, pathology is often supported by decrease in intraocular pigment epithelium-derived factor (PEDF), an endogenous inhibitor of angiogenesis. A surface helical 34-mer peptide of PEDF, comprising this activity, is efficacious in animal models of neovascular retina disease but remains impractically large for therapeutic use. We sought smaller fragments within this sequence that mitigate choroidal neovascularization (CNV). Expecting rapid intravitreal (IVT) clearance, we also developed a method to reversibly attach peptides to nano-carriers for extended delivery. Synthetic fragments of 34-mer yielded smaller anti-angiogenic peptides, and N-terminal capping with dicarboxylic acids did not diminish activity. Charge restoration via substitution of an internal aspartate by asparagine improved potency, achieving low nM apoptotic response in VEGF-activated EC. Two optimized peptides (PEDF 335, 8-mer and PEDF 336, 9-mer) were tested in a mouse model of laser-induced CNV. IVT injection of either peptide, 2-5 days before laser treatment, gave significant CNV decrease at day +14 post laser treatment. The 8-mer also decreased CNV, when administered as eye drops. Also examined was a nanoparticle-conjugate (NPC) prodrug of the 9-mer, having positive zeta potential, expected to display longer intraocular residence. This NPC showed extended efficacy, even when injected 14 days before laser treatment. Neither inflammatory cells nor other histopathologic abnormalities were seen in rabbit eyes harvested 14 days following IVT injection of PEDF 336 (>200 µg). No rabbit or mouse eye irritation was observed over 12-17 days of PEDF 335 eye drops (10 mM). Viability was unaffected in 3 retinal and 2 choroidal cell types by PEDF 335 up to 100 µM, PEDF 336 (100 µM) gave slight growth inhibition only in choroidal EC. A small anti-angiogenic PEDF epitope (G-Y-D-L-Y-R-V) was identified, variants (adipic-Sar-Y-N-L-Y-R-V) mitigate CNV, with clinical potential in treating neovascular retinopathy. Their shared active motif, Y - - - R, is found in laminin (Ln) peptide YIGSR, which binds Ln receptor 67LR, a known high-affinity ligand of PEDF 34-mer.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Choroidal Neovascularization/prevention & control , Eye Proteins/therapeutic use , Nerve Growth Factors/therapeutic use , Oligopeptides/therapeutic use , Serpins/therapeutic use , Administration, Ophthalmic , Angiogenesis Inhibitors/chemistry , Animals , Apoptosis , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Disease Models, Animal , Drug Carriers , Electroretinography , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Eye Proteins/chemistry , Mice , Mice, Inbred C57BL , Nerve Growth Factors/chemistry , Oligopeptides/chemistry , Ophthalmic Solutions , Prodrugs , Rabbits , Rats , Serpins/chemistry
4.
Neoplasia ; 20(9): 930-942, 2018 09.
Article in English | MEDLINE | ID: mdl-30118999

ABSTRACT

We have previously demonstrated that apigenin promotes the expression of antiangiogenic protein thrombospondin-1 (TSP1) via a mechanism driven by mRNA-binding protein HuR. Here, we generated a novel mouse model with whole-body THBS-1 gene knockout on SKH-1 genetic background, which allows studies of UVB-induced acute skin damage and carcinogenesis and tests TSP1 involvement in apigenin's anticancer effects. Apigenin significantly inhibited UVB-induced carcinogenesis in the wild-type (WT) animals but not in TSP1 KO (TKO) mice, suggesting that TSP1 is a critical component of apigenin's chemopreventive function in UVB-induced skin cancer. Importantly, TKO mice presented with the elevated cutaneous inflammation at baseline, which was manifested by increased inflammatory infiltrates (neutrophils and macrophages) and elevated levels of the two key inflammatory cytokines, IL-6 and IL-12. In agreement, maintaining normal TSP1 expression in the UVB-irradiated skin of WT mice using topical apigenin application caused a marked decrease of circulating inflammatory cytokines. Finally, TKO mice showed an altered population dynamics of the bone marrow myeloid progenitor cells (CD11b+), with dramatic expansion of the population of neutrophil progenitors (Ly6ClowLy6Ghigh) compared to the WT control. Our results indicate that the cutaneous tumor suppressor TSP1 is a critical mediator of the in vivo anticancer effect of apigenin in skin, specifically of its anti-inflammatory action.


Subject(s)
Apigenin/pharmacology , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/radiation effects , Skin Neoplasms/etiology , Skin Neoplasms/pathology , Sunscreening Agents/pharmacology , Ultraviolet Rays/adverse effects , Animals , Anti-Inflammatory Agents , Cell Line, Tumor , Chemoprevention , Disease Models, Animal , Genotype , Humans , Inflammation/etiology , Inflammation/pathology , Inflammation/prevention & control , Keratinocytes/drug effects , Keratinocytes/metabolism , Mice , Mice, Knockout , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Peroxidase/metabolism , Skin/drug effects , Skin/metabolism , Skin/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/prevention & control , Thrombospondin 1/genetics , Thrombospondin 1/metabolism , Xenograft Model Antitumor Assays
5.
Nat Commun ; 8(1): 1319, 2017 11 06.
Article in English | MEDLINE | ID: mdl-29105655

ABSTRACT

Metastatic cancers produce exosomes that condition pre-metastatic niches in remote microenvironments to favor metastasis. In contrast, here we show that exosomes from poorly metastatic melanoma cells can potently inhibit metastasis to the lung. These "non-metastatic" exosomes stimulate an innate immune response through the expansion of Ly6Clow patrolling monocytes (PMo) in the bone marrow, which then cause cancer cell clearance at the pre-metastatic niche, via the recruitment of NK cells and TRAIL-dependent killing of melanoma cells by macrophages. These events require the induction of the Nr4a1 transcription factor and are dependent on pigment epithelium-derived factor (PEDF) on the outer surface of exosomes. Importantly, exosomes isolated from patients with non-metastatic primary melanomas have a similar ability to suppress lung metastasis. This study thus demonstrates that pre-metastatic tumors produce exosomes, which elicit a broad range of PMo-reliant innate immune responses via trigger(s) of immune surveillance, causing cancer cell clearance at the pre-metastatic niche.


Subject(s)
Exosomes/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/secondary , Monocytes/immunology , Animals , Cell Differentiation/immunology , Eye Proteins/immunology , Female , Humans , Immunity, Innate , Immunologic Surveillance , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Macrophages/immunology , Macrophages/pathology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Nude , Monocytes/pathology , Nerve Growth Factors/immunology , Phagocytosis/immunology , Serpins/immunology , Tumor Microenvironment/immunology
6.
PLoS One ; 12(4): e0174968, 2017.
Article in English | MEDLINE | ID: mdl-28403150

ABSTRACT

BACKGROUND: Although inflammation and prostate cancer (PCa) have been linked, the molecular interactions between macrophages and PCa cells are poorly explored. Pigment Epithelium-Derived Factor (PEDF) is an anti-angiogenic and anti-tumor factor. We previously showed that PEDF induces macrophages recruitment in vitro, correlates with macrophages density in human prostate, and stimulates macrophages polarization towards the classically activated pathway. Here, we demonstrate that PEDF modulates the interaction between macrophages and PCa cells through a bidirectional signalling leading to tumor cell apoptosis and phagocytosis. METHODS: RAW 264.7 and THP-1 cells, and BMDMs were grown in vitro as mono- or co-cultures with PC3 or CL1 tumor cells. The effects of PEDF and its derived P18 peptide were measured on macrophages differentiation, migration, and superoxide production, and tumor cell apoptosis and phagocytosis. PEDF receptors (ATP5B, PNPLA2, and LRP6) and CD47 mRNA and protein expression were quantified in macrophages and tumor cells by quantitative RT-PCR, western blot, immunofluorescence and flow cytometry. RESULTS: We found that PEDF induced the migration of macrophages towards tumor 3D spheroids and 2D cultures. In co-culture, PEDF increased PCa cells phagocytosis through an indirect apoptosis-dependent mechanism. Moreover, PEDF stimulated the production of superoxide by macrophages. Conditioned media from macrophages exposed to PEDF induced tumor cells apoptosis in contrast to control conditioned media suggesting that ROS may be involved in tumor cells apoptosis. ATP5B and PNPLA2 PEDF receptors on macrophages and CD47 on tumor cells were respectively up- and down-regulated by PEDF. As PEDF, blocking CD47 induced phagocytosis. Inhibiting ATP5B reduced phagocytosis. Inversely, PNPLA2 inhibition blocks differentiation but maintains phagocytosis. CD47-induced phagocytosis was partially reverted by ATP5B inhibition suggesting a complementary action. Similar effects were observed with P18 PEDF-derived peptide. CONCLUSIONS: These data established that modulating the molecular interactions between macrophages and PCa cells using PEDF may be a promising strategy for PCa treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Eye Proteins/pharmacology , Macrophages/physiology , Nerve Growth Factors/pharmacology , Prostatic Neoplasms/drug therapy , Serpins/pharmacology , Animals , CD47 Antigen/metabolism , Cell Line, Tumor , Cell Movement , Coculture Techniques , Drug Screening Assays, Antitumor , Humans , Lipase/metabolism , Macrophages/drug effects , Male , Mice , Mitochondrial Proton-Translocating ATPases/metabolism , Phagocytosis , Phenylurea Compounds/pharmacology , RAW 264.7 Cells , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Superoxides/metabolism
7.
Sci Rep ; 6: 22915, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-26964503

ABSTRACT

Exosomes are produced by cells to mediate intercellular communication, and have been shown to perpetuate diseases, including cancer. New tools are needed to understand exosome biology, detect exosomes from specific cell types in complex biological media, and to modify exosomes. Our data demonstrate a cellular pathway whereby membrane-bound scavenger receptor type B-1 (SR-B1) in parent cells becomes incorporated into exosomes. We tailored synthetic HDL-like nanoparticles (HDL NP), high-affinity ligands for SR-B1, to carry a fluorescently labeled phospholipid. Data show SR-B1-dependent transfer of the fluorescent phospholipid from HDL NPs to exosomes. Modified exosomes are stable in serum and can be directly detected using flow cytometry. As proof-of-concept, human serum exosomes were found to express SR-B1, and HDL NPs can be used to label and isolate them. Ultimately, we discovered a natural cellular pathway and nanoparticle-receptor pair that enables exosome modulation, detection, and isolation.


Subject(s)
Biosensing Techniques , Cell Communication/genetics , Exosomes/metabolism , Scavenger Receptors, Class B/isolation & purification , Exosomes/chemistry , Humans , Ligands , Lipid Metabolism/genetics , Lipoproteins, HDL/chemistry , Nanoparticles/chemistry , Phospholipids/chemistry , Phospholipids/metabolism , Protein Binding , Scavenger Receptors, Class B/blood , Scavenger Receptors, Class B/chemistry , Scavenger Receptors, Class B/genetics
8.
Cell Signal ; 28(5): 460-468, 2016 May.
Article in English | MEDLINE | ID: mdl-26876613

ABSTRACT

Ultraviolet B (UVB) radiation is the major environmental risk factor for developing skin cancer, the most common cancer worldwide, which is characterized by aberrant activation of Akt/mTOR (mammalian target of rapamycin). Importantly, the link between UV irradiation and mTOR signaling has not been fully established. Apigenin is a naturally occurring flavonoid that has been shown to inhibit UV-induced skin cancer. Previously, we have demonstrated that apigenin activates AMP-activated protein kinase (AMPK), which leads to suppression of basal mTOR activity in cultured keratinocytes. Here, we demonstrated that apigenin inhibited UVB-induced mTOR activation, cell proliferation and cell cycle progression in mouse skin and in mouse epidermal keratinocytes. Interestingly, UVB induced mTOR signaling via PI3K/Akt pathway, however, the inhibition of UVB-induced mTOR signaling by apigenin was not Akt-dependent. Instead, it was driven by AMPK activation. In addition, mTOR inhibition by apigenin in keratinocytes enhanced autophagy, which was responsible, at least in part, for the decreased proliferation in keratinocytes. In contrast, apigenin did not alter UVB-induced apoptosis. Taken together, our results indicate the important role of mTOR inhibition in UVB protection by apigenin, and provide a new target and strategy for better prevention of UV-induced skin cancer.


Subject(s)
Anticarcinogenic Agents/pharmacology , Apigenin/pharmacology , Keratinocytes/drug effects , Keratinocytes/radiation effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Ultraviolet Rays , AMP-Activated Protein Kinases/metabolism , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Cycle/drug effects , Cell Line , Cell Proliferation/drug effects , Keratinocytes/cytology , Keratinocytes/enzymology , Mice, Inbred BALB C , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/radiation effects , Skin Neoplasms/prevention & control , TOR Serine-Threonine Kinases/metabolism
9.
Cancer Res ; 76(8): 2265-76, 2016 04 15.
Article in English | MEDLINE | ID: mdl-26921338

ABSTRACT

Loss of pigment epithelium-derived factor (PEDF, SERPINF1) in cancer cells is associated with poor prognosis and metastasis, but the contribution of stromal PEDF to cancer evolution is poorly understood. Therefore, we investigated the role of fibroblast-derived PEDF in melanoma progression. We demonstrate that normal dermal fibroblasts expressing high PEDF levels attenuated melanoma growth and angiogenesis in vivo, whereas PEDF-depleted fibroblasts exerted tumor-promoting effects. Accordingly, mice with global PEDF knockout were more susceptible to melanoma metastasis. We also demonstrate that normal fibroblasts in close contact with PEDF-null melanoma cells lost PEDF expression and tumor-suppressive properties. Further mechanistic investigations underlying the crosstalk between tumor and stromal cells revealed that melanoma cells produced PDGF-BB and TGFß, which blocked PEDF production in fibroblasts. Notably, cancer-associated fibroblasts (CAF) isolated from patient-derived tumors expressed markedly low levels of PEDF. Treatment of patient CAF and TGFß-treated normal fibroblasts with exogenous PEDF decreased the expression of CAF markers and restored PEDF expression. Finally, expression profiling of PEDF-depleted fibroblasts revealed induction of IL8, SERPINB2, hyaluronan synthase-2, and other genes associated with tumor promotion and metastasis. Collectively, our results demonstrate that PEDF maintains tumor-suppressive functions in fibroblasts to prevent CAF conversion and illustrate the mechanisms by which melanoma cells silence stromal PEDF to promote malignancy. Cancer Res; 76(8); 2265-76. ©2016 AACR.


Subject(s)
Cell Proliferation , Eye Proteins/biosynthesis , Fibroblasts/metabolism , Melanoma/pathology , Nerve Growth Factors/biosynthesis , Serpins/biosynthesis , Animals , Cell Line, Tumor , Humans , Melanoma/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Transforming Growth Factor beta/metabolism
10.
Vasc Cell ; 7: 6, 2015.
Article in English | MEDLINE | ID: mdl-26161255

ABSTRACT

BACKGROUND: Multiple studies demonstrated pro-angiogenic effects of microRNA (miR)-27b. Its targets include Notch ligand Dll4, Sprouty (Spry)-2, PPARγ and Semaphorin (SEMA) 6A. miR-27 effects in the heart are context-dependent: although it is necessary for ventricular maturation, targeted overexpression in cardiomyocytes causes hypertrophy and dysfunction during development. Despite significant recent advances, therapeutic potential of miR-27b in cardiovascular disease and its effects in adult heart remain unexplored. Here, we assessed the therapeutic potential of miR-27b mimics and inhibitors in rodent models of ischemic disease and cancer. METHODS: We have used a number of models to demonstrate the effects of miR-27b mimicry and inhibition in vivo, including subcutaneous Matrigel plug assay, mouse models of hind limb ischemia and myocardial infarction and subcutaneous Lewis Lung carcinoma. RESULTS: Using mouse model of myocardial infarction due to the coronary artery ligation, we showed that miR-27b mimic had overall beneficial effects, including increased vascularization, decreased fibrosis and increased ejection fraction. In mouse model of critical limb ischemia, miR-27b mimic also improved tissue re-vascularization and perfusion. In both models, miR-27b mimic clearly decreased macrophage recruitment to the site of hypoxic injury. In contrast, miR-27b increased the recruitment of bone marrow derived cells to the neovasculature, as was shown using mice reconstituted with fluorescence-tagged bone marrow. These effects were due, at least in part, to the decreased expression of Dll4, PPARγ and IL10. In contrast, blocking miR-27b significantly decreased vascularization and reduced growth of subcutaneous tumors and decreased BMDCs recruitment to the tumor vasculature. CONCLUSIONS: Our study demonstrates the utility of manipulating miR-27b levels in the treatment of cardiovascular disease and cancer.

11.
Oncotarget ; 5(22): 11413-27, 2014 Nov 30.
Article in English | MEDLINE | ID: mdl-25526033

ABSTRACT

Plant flavonoid apigenin prevents and inhibits UVB-induced carcinogenesis in the skin and has strong anti-proliferative and anti-angiogenic properties. Here we identify mechanisms, by which apigenin controls these oncogenic events. We show that apigenin acts, at least in part, via endogenous angiogenesis inhibitor, thrombospondin-1 (TSP1). TSP1 expression by the epidermal keratinocytes is potently inhibited by UVB. It inhibits cutaneous angiogenesis and UVB-induced carcinogenesis. We show that apigenin restores TSP1 in epidermal keratinocytes subjected to UVB and normalizes proliferation and angiogenesis in UVB-exposed skin. Importantly, reconstituting TSP1 anti-angiogenic function in UVB-irradiated skin with a short bioactive peptide mimetic representing exclusively its anti-angiogenic domain reproduced the anti-proliferative and anti-angiogenic effects of apigenin. Cox-2 and HIF-1α are important mediators of angiogenesis. Both apigenin and TSP1 peptide mimetic attenuated their induction by UVB. Finally we identified the molecular mechanism, whereby apigenin did not affect TSP1 mRNA, but increased de novo protein synthesis. Knockdown studies implicated the RNA-binding protein HuR, which controls mRNA stability and translation. Apigenin increased HuR cytoplasmic localization and physical association with TSP1 mRNA causing de novo TSP1 synthesis. HuR cytoplasmic localization was, in turn, dependent on CHK2 kinase. Together, our data provide a new mechanism, by which apigenin controls UVB-induced carcinogenesis.


Subject(s)
Apigenin/pharmacology , ELAV Proteins/metabolism , Keratinocytes/drug effects , Keratinocytes/radiation effects , Neoplasms, Radiation-Induced/prevention & control , Skin Neoplasms/prevention & control , Skin/drug effects , Skin/radiation effects , Thrombospondin 1/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Cell Growth Processes/drug effects , Cell Growth Processes/radiation effects , Chemoprevention/methods , Female , Humans , Keratinocytes/metabolism , Mice , Mice, Hairless , Mice, Inbred BALB C , Neoplasms, Radiation-Induced/blood supply , Neoplasms, Radiation-Induced/metabolism , Neovascularization, Pathologic , Skin/blood supply , Skin/metabolism , Skin Neoplasms/blood supply , Skin Neoplasms/metabolism , Ultraviolet Rays
12.
Part Part Syst Charact ; 31(11): 1141-1150, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25400330

ABSTRACT

Systemic delivery of therapeutic nucleic acids to target cells and tissues outside of the liver remains a major challenge. We synthesized a biomimetic high density lipoprotein nanoparticle (HDL NP) for delivery of a cholesteryl modified therapeutic nucleic acid (RNAi) to vascular endothelial cells, a cell type naturally targeted by HDL. HDL NPs adsorb cholesteryl modified oligonucleotides and protect them from nuclease degradation. As proof of principle, we delivered RNAi targeting vascular endothelial growth factor receptor 2 (VEGFR2) to endothelial cells to effectively silence target mRNA and protein expression in vitro. In addition, data show that treatment strongly attenuated in vivo neovascularization measured using a standard angiogenesis assay and in hypervascular tumor allografts where a striking reduction in tumor growth was observed. For effective delivery, HDL NPs required the expression of the cell surface protein scavenger receptor type-B1 (SR-B1). No toxicity of HDL NPs was measured in vitro or after in vivo administration. Thus, by using a biomimetic approach to nucleic acid delivery, data demonstrate that systemically administered RNAi-HDL NPs target SR-B1 expressing endothelial cells to deliver functional anti-angiogenic RNAi as a potential treatment of cancer and other neo-vascular diseases.

13.
Expert Opin Drug Discov ; 8(7): 769-92, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23642051

ABSTRACT

INTRODUCTION: Pigment epithelium-derived factor (PEDF) was discovered as a neurotrophic factor secreted by retinal pigment epithelial cells. A decade later, it re-emerged as a powerful angiogenesis inhibitor guarding ocular function. Since then, significant advances were made identifying PEDF's mechanisms, targets and biomedical applications. AREAS COVERED: The authors review several methodologies that have generated significant new information about the potential of PEDF as a drug. Furthermore, the authors review and discuss mechanistic and structure-function analyses combined with the functional mapping of active fragments, which have yielded several short bioactive PEDF peptides. Additionally, the authors present functional studies in knockout animals and human correlates that have provided important information about conditions amenable to PEDF-based therapies. EXPERT OPINION: Through its four known receptors, PEDF causes a wide range of cellular events vitally important for the organism, which include survival and differentiation, migration and invasion, lipid metabolism and stem cell maintenance. These processes are deregulated in multiple pathological conditions, including cancer, metabolic and cardiovascular disease. PEDF has been successfully used in countless preclinical models of these conditions and human correlates suggest a wide utility of PEDF-based drugs. The most significant clinical application of PEDF, to date, is its potential therapeutic use for age-related macular degeneration. Moreover, PEDF-based gene therapy has advanced to early stage clinical trials. PEDF active fragments have been mapped and used to design short peptide mimetics conferring distinct functions of PEDF, which may address specific clinical problems and become prototype drugs.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Eye Proteins/therapeutic use , Lipid Metabolism/drug effects , Macular Degeneration/drug therapy , Nerve Growth Factors/therapeutic use , Protease Inhibitors/therapeutic use , Serpins/therapeutic use , Animals , Cardiovascular Diseases/drug therapy , Clinical Trials as Topic , Drug Discovery , Humans , Metabolic Diseases/drug therapy , Models, Animal , Neoplasms/drug therapy
14.
PLoS One ; 8(12): e83991, 2013.
Article in English | MEDLINE | ID: mdl-24391862

ABSTRACT

miRNA regulate gene expression at post-transcriptional level and fine-tune the key biological processes, including cancer progression. Here, we demonstrate the involvement of miR-200 b in the metastatic spread of prostate cancer. We identified miR-200 b as a downstream target of androgen receptor and linked its expression to decreased tumorigenicity and metastatic capacity of the prostate cancer cells. Overexpression of miR-200 b in PC-3 cells significantly inhibited their proliferation and the formation of subcutaneous tumors. Moreover, in an orthotopic model, miR-200 b blocked spontaneous metastasis and angiogenesis by PC-3 cells. This decreased metastatic potential was likely due to the reversal of the epithelial-to-mesenchymal transition, as was evidenced by increased pan-epithelial marker E-cadherin and specific markers of prostate epithelium, cytokeratins 8 and 18. In contrast, mesenchymal markers, fibronectin and vimentin, were significantly downregulated by miR-200 b. Our results suggest an important role for miR-200 b in prostate cancer progression and indicate its potential utility for prostate cancer therapy.


Subject(s)
Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , MicroRNAs/genetics , Prostatic Neoplasms/pathology , Animals , Apoptosis , Blotting, Western , Cadherins/genetics , Cadherins/metabolism , Cell Cycle , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Male , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/secondary , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Vimentin/genetics , Vimentin/metabolism , Xenograft Model Antitumor Assays
15.
FASEB J ; 26(11): 4685-95, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22874833

ABSTRACT

Sucrose nonfermenting 1 (Snf1)-related kinase (SNRK) is a serine/threonine kinase with sequence similarity to AMP-activated protein kinase (AMPK); however, its function is not well characterized. We conducted a gene array to determine which genes are regulated by SNRK. The array demonstrated that SNRK overexpression increased the levels of genes involved in cell proliferation, including calcyclin-binding protein (CacyBP), a member of the ubiquitin ligase complex that targets nonphosphorylated ß-catenin for degradation. We confirmed that SNRK increased CacyBP mRNA and protein, and decreased ß-catenin protein in HCT116 and RKO colon cancer cells. Furthermore, SNRK inhibited colon cancer cell proliferation, and CacyBP down-regulation reversed the SNRK-mediated decrease in proliferation and ß-catenin. SNRK overexpression also decreased ß-catenin nuclear localization and target gene transcription, and ß-catenin down-regulation reversed the effects of SNRK knockdown on proliferation. SNRK transcript levels were reduced in human colon tumors compared to normal tissue by 35.82%, and stable knockdown of SNRK increased colon cancer cell tumorigenicity. Our results demonstrate that SNRK is down-regulated in colon cancer and inhibits colon cancer cell proliferation through CacyBP up-regulation and ß-catenin degradation, resulting in reduced proliferation signaling. These findings reveal a novel function for SNRK in the regulation of colon cancer cell proliferation and ß-catenin signaling.


Subject(s)
Calcium-Binding Proteins/metabolism , Colonic Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , beta Catenin/metabolism , Calcium-Binding Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic/physiology , Gene Knockdown Techniques , Humans , Protein Serine-Threonine Kinases/genetics , Signal Transduction/physiology , Transcriptome , beta Catenin/genetics
16.
PLoS One ; 7(3): e31052, 2012.
Article in English | MEDLINE | ID: mdl-22403609

ABSTRACT

The accepted androgen receptor (AR) role is to promote proliferation and survival of prostate epithelium and thus prostate cancer progression. While growth-inhibitory, tumor-suppressive AR effects have also been documented, the underlying mechanisms are poorly understood. Here, we for the first time link AR anti-cancer action with cell senescence in vitro and in vivo. First, AR-driven senescence was p53-independent. Instead, AR induced p21, which subsequently reduced ΔN isoform of p63. Second, AR activation increased reactive oxygen species (ROS) and thereby suppressed Rb phosphorylation. Both pathways were critical for senescence as was proven by p21 and Rb knock-down and by quenching ROS with N-Acetyl cysteine and p63 silencing also mimicked AR-induced senescence. The two pathways engaged in a cross-talk, likely via PML tumor suppressor, whose localization to senescence-associated chromatin foci was increased by AR activation. All these pathways contributed to growth arrest, which resolved in senescence due to concomitant lack of p53 and high mTOR activity. This is the first demonstration of senescence response caused by a nuclear hormone receptor.


Subject(s)
Cellular Senescence , Receptors, Androgen/metabolism , Androgen Receptor Antagonists/pharmacology , Cell Line, Tumor , Cellular Senescence/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytokines/metabolism , Flutamide/pharmacology , Humans , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , Phosphorylation/drug effects , Promyelocytic Leukemia Protein , Reactive Oxygen Species/metabolism , Retinoblastoma Protein/metabolism , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Up-Regulation/drug effects
17.
Blood ; 119(11): 2679-87, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22207734

ABSTRACT

We discovered that miR-27b controls 2 critical vascular functions: it turns the angiogenic switch on by promoting endothelial tip cell fate and sprouting and it promotes venous differentiation. We have identified its targets, a Notch ligand Delta-like ligand 4 (Dll4) and Sprouty homologue 2 (Spry2). miR-27b knockdown in zebrafish and mouse tissues severely impaired vessel sprouting and filopodia formation. Moreover, miR-27b was necessary for the formation of the first embryonic vein in fish and controlled the expression of arterial and venous markers in human endothelium, including Ephrin B2 (EphB2), EphB4, FMS-related tyrosine kinase 1 (Flt1), and Flt4. In zebrafish, Dll4 inhibition caused increased sprouting and longer intersegmental vessels and exacerbated tip cell migration. Blocking Spry2 caused premature vessel branching. In contrast, Spry2 overexpression eliminated the tip cell branching in the intersegmental vessels. Blockade of Dll4 and Spry2 disrupted arterial specification and augmented the expression of venous markers. Blocking either Spry2 or Dll4 rescued the miR-27b knockdown phenotype in zebrafish and in mouse vascular explants, pointing to essential roles of these targets downstream of miR-27b. Our study identifies critical role of miR-27b in the control of endothelial tip cell fate, branching, and venous specification and determines Spry2 and Dll4 as its essential targets.


Subject(s)
Arteries/embryology , Endothelium, Vascular/cytology , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , MicroRNAs/physiology , Neovascularization, Physiologic , Zebrafish/genetics , Adaptor Proteins, Signal Transducing , Animals , Aorta/cytology , Aorta/metabolism , Arteries/metabolism , Biomarkers/metabolism , Blotting, Northern , Blotting, Western , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Cell Differentiation , Cell Movement , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Endothelium, Vascular/metabolism , Gene Expression Profiling , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Oligonucleotide Array Sequence Analysis , Protein Serine-Threonine Kinases , Pseudopodia/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Signal Transduction , Zebrafish/embryology , Zebrafish/metabolism
18.
Expert Opin Ther Targets ; 15(12): 1369-86, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22136063

ABSTRACT

INTRODUCTION: The role of hrombospondin-1 (TSP1) as a major endogenous angiogenesis inhibitor has been confirmed by numerous studies and subsequent mechanistic discoveries. It has yielded a new class of potential drugs against cancer and other angiogenesis-driven diseases. AREAS COVERED: An overview of TSP1 functions and molecular mechanisms, including regulation and signaling. Functions in endothelial and non-endothelial cells, with emphasis on the role of TSP1 in the regulation of angiogenesis and inflammation. The utility of duplicating these activities for drug discovery. Past and current literature on endogenous TSP1 and its role in the progression of cancer and non-cancerous pathological conditions is summarized, as well as the research undertaken to identify and optimize short bioactive peptides derived from the two TSP1 anti-angiogenic domains, which bind CD47 and CD36 cell surface receptors. Lastly, there is an overview of the efficacy of some of these peptides in pre-clinical and clinical models of angiogenesis-dependent disease. EXPERT OPINION: It is concluded that TSP1-derived peptides and peptide mimetics hold great promise as future agents for the treatment of cancer and other diseases driven by excessive angiogenesis. They may fulfill unmet medical needs including neovascular ocular disease and the diseases of the female reproductive tract including ovarian cancer.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Neovascularization, Pathologic/drug therapy , Peptides/therapeutic use , Thrombospondin 1/metabolism , Animals , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Receptors, Cell Surface , Signal Transduction
19.
Cancer Res ; 71(24): 7490-501, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22028325

ABSTRACT

Thrombospondin-1 (TSP-1) is an endogenous inhibitor of angiogenesis encoded by the THBS1 gene, whose promoter is activated by p53. In advanced colorectal cancers (CRC), its expression is sustained or even slightly increased despite frequent loss of p53. Here, we determined that in HCT116 CRC cells, p53 activates the THBS1 primary transcript, but fails to boost THBS1 mRNA or protein levels, implying posttranscriptional regulation by microRNAs (miRNA). In a global miRNA gain-of-function screen done in the Dicer-deficient HCT116 variant, several miRNAs negatively regulated THBS1 mRNA and protein levels, one of them being miR-194. Notably, in agreement with published data, p53 upregulated miR-194 expression in THBS1 retrovirus-transduced HCT116 cells, leading to decreased TSP-1 levels. This negative effect was mediated by a single miR-194 complementary site in the THBS1 3'-untranslated region, and its elimination resulted in TSP-1 reactivation, impaired angiogenesis in Matrigel plugs, and reduced growth of HCT116 xenografts. Conversely, transient overexpression of miR-194 in HCT116/THBS1 cells boosted Matrigel angiogenesis, and its stable overexpression in Ras-induced murine colon carcinomas increased microvascular densities and vessel sizes. Although the overall contribution of miR-194 to neoplastic growth is context dependent, p53-induced activation of this GI tract-specific miRNA during ischemia could promote angiogenesis and facilitate tissue repair.


Subject(s)
Colonic Neoplasms/genetics , MicroRNAs/genetics , Neovascularization, Pathologic/genetics , Thrombospondin 1/genetics , Tumor Suppressor Protein p53/genetics , 3' Untranslated Regions/genetics , Animals , Blotting, Western , Cell Line, Tumor , Colonic Neoplasms/blood supply , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Mice , MicroRNAs/metabolism , Mutation , Retroviridae/genetics , Reverse Transcriptase Polymerase Chain Reaction , Thrombospondin 1/metabolism , Transcription, Genetic , Transduction, Genetic , Transplantation, Heterologous , Tumor Suppressor Protein p53/metabolism
20.
Neoplasia ; 13(7): 633-42, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21750657

ABSTRACT

Metastatic melanoma cells are highly adaptable to their in vivo microenvironment and can switch between protease-dependent mesenchymal and protease-independent amoeboid invasion to facilitate metastasis. Such adaptability can be visualized in vitro, when cells are cultured in conditions that recapitulate three-dimensional microenvironments. Using thick collagen layers in cell culture and in vivo extravasation assays, we found that pigment epithelium-derived factor (PEDF) suppressed lung extravasation of aggressive melanoma by coordinated regulation of cell shape and proteolysis. In cells grown on a thick collagen bed, PEDF overexpression and exogenous PEDF blocked the rapidly invasive, rounded morphology, and promoted an elongated, mesenchymal-like phenotype associated with reduced invasion. These changes in cell shape depended on decreased RhoA and increased Rac1 activation and were mediated by the up-regulation of Rac1-GEF, DOCK3 and down-regulation of Rac1-GAP, ARHGAP22. Surprisingly, we found that PEDF overexpression also blocked the trafficking of membrane-tethered, MT1-MMP to the cell surface through RhoA inhibition and Rac1 activation. In vivo, knockdown of Rac1 and DOCK3 or overexpression of MT1-MMP was sufficient to reverse the inhibitory effect of PEDF on extravasation. Using functional studies, we demonstrated that PEDF suppressed the rounded morphology and MT1-MMP surface localization through its antiangiongenic, 34-mer epitope and the recently identified PEDF receptor candidate, PNPLA2. Our findings unveil the coordinated regulation of cell shape and proteolysis and identify an unknown mechanism for PEDF's antimetastatic activity.


Subject(s)
Amoeba/cytology , Eye Proteins/pharmacology , Mesoderm/drug effects , Neoplasm Invasiveness/prevention & control , Neoplasms/pathology , Nerve Growth Factors/pharmacology , Protein Processing, Post-Translational/drug effects , Pseudopodia/drug effects , Serpins/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Shape/drug effects , Cells, Cultured , Epithelial-Mesenchymal Transition/drug effects , Eye Proteins/therapeutic use , Humans , Mesoderm/metabolism , Mice , Mice, Nude , Neoplasms/drug therapy , Nerve Growth Factors/therapeutic use , Pseudopodia/pathology , Serpins/therapeutic use , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...