Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 123
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 119(45): e2204795119, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36322719

ABSTRACT

Abnormal neovascularization is an important cause of blindness in many ocular diseases, for which the etiology and pathogenic mechanisms remain incompletely understood. Recent studies have revealed the diverse roles of noncoding RNAs in various biological processes and facilitated the research and development of the clinical application of numerous RNA drugs, including microRNAs. Here, we report the antiangiogenic activity of microRNA-29a (miR-29a) in three animal models of ocular neovascularization. The miR-29a knockout (KO) mice displayed enhanced vessel pruning, resulting in a decreased vascularized area during retinal development. In contrast, miR-29a deletion in adult mice accelerated angiogenesis in preclinical disease models, including corneal neovascularization, oxygen-induced retinopathy, and choroidal neovascularization, while the administration of agomir-29a ameliorated pathological neovascularization. Furthermore, miR-29a exerted inhibitory effects on endothelial cell proliferation, migration, and tube formation capacities. RNA sequencing analysis of retinas from miR-29a KO mice and RNA interference experiments identified platelet-derived growth factor C and several extracellular matrix genes as downstream targets of miR-29a involved in regulating ocular angiogenesis. Our data suggest that miR-29a may be a promising clinical candidate for the treatment of neovascular diseases.


Subject(s)
Choroidal Neovascularization , MicroRNAs , Mice , Animals , MicroRNAs/metabolism , Cell Proliferation , RNA Interference , Eye/metabolism , Choroidal Neovascularization/metabolism , Mice, Knockout
2.
FASEB J ; 35(5): e21404, 2021 05.
Article in English | MEDLINE | ID: mdl-33899275

ABSTRACT

We have previously used the genetic diversity available in common inbred mouse strains to identify quantitative trait loci (QTLs) responsible for the differences in angiogenic response using the corneal micropocket neovascularization (CoNV) assay. Employing a mouse genome-wide association study (GWAS) approach, the region on chromosome 15 containing Basp1 was identified as being significantly associated with angiogenesis in inbred strains. Here, we developed a unique strategy to determine and verify the role of BASP1 in angiogenic pathways. Basp1 expression in cornea had a strong correlation with a haplotype shared by mouse strains with varied angiogenic phenotypes. In addition, inhibition of BASP1 demonstrated a dosage-dependent effect in both primary mouse brain endothelial and human microvascular endothelial cell (HMVEC) migration. To investigate its role in vivo, we knocked out basp1 in transgenic kdrl:zsGreen zebrafish embryos using a widely adopted CRISPR-Cas9 system. These embryos had severely disrupted vessel formation compared to control siblings. We further show that basp1 promotes angiogenesis by upregulating ß-catenin gene and the Dll4/Notch1 signaling pathway. These results, to the best of our knowledge, provide the first in vivo evidence to indicate the role of Basp1 as an angiogenesis-regulating gene and opens the potential therapeutic avenues for a wide variety of systemic angiogenesis-dependent diseases.


Subject(s)
Corneal Neovascularization/pathology , Membrane Proteins/metabolism , Models, Biological , Neovascularization, Pathologic/pathology , Nerve Tissue Proteins/metabolism , Repressor Proteins/metabolism , Animals , Cell Movement , Corneal Neovascularization/genetics , Corneal Neovascularization/metabolism , Genome-Wide Association Study , Humans , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Morphogenesis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Nerve Tissue Proteins/genetics , Repressor Proteins/genetics , Wnt Signaling Pathway , Zebrafish
3.
Angiogenesis ; 24(1): 111-127, 2021 02.
Article in English | MEDLINE | ID: mdl-32955682

ABSTRACT

Angiogenesis plays a key role in the pathology of diseases such as cancer, diabetic retinopathy, and age-related macular degeneration. Understanding the driving forces of endothelial cell migration and organization, as well as the time frame of these processes, can elucidate mechanisms of action of important pathological pathways. Herein, we have developed an organ-specific microfluidic platform recapitulating the in vivo angiogenic microenvironment by co-culturing mouse primary brain endothelial cells with brain pericytes in a three-dimensional (3D) collagen scaffold. As a proof of concept, we show that this model can be used for studying the angiogenic process and further comparing the angiogenic properties between two different common inbred mouse strains, C57BL/6J and 129S1/SvlmJ. We further show that the newly discovered angiogenesis-regulating gene Padi2 promotes angiogenesis through Dll4/Notch1 signaling by an on-chip mechanistic study. Analysis of the interplay between primary endothelial cells and pericytes in a 3D microfluidic environment assists in the elucidation of the angiogenic response.


Subject(s)
Cell Engineering , Cellular Microenvironment , Endothelial Cells/pathology , Imaging, Three-Dimensional , Microfluidics , Pericytes/pathology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Calcium-Binding Proteins/metabolism , Cell Separation , Cells, Cultured , Down-Regulation , Endothelial Cells/metabolism , Mice, Inbred C57BL , Neovascularization, Pathologic/pathology , Pericytes/metabolism , Protein-Arginine Deiminase Type 2/antagonists & inhibitors , Protein-Arginine Deiminase Type 2/metabolism , Receptors, Notch/metabolism , Signal Transduction
4.
Int J Mol Sci ; 22(10)2021 May 13.
Article in English | MEDLINE | ID: mdl-34068392

ABSTRACT

Myocardial infarction (MI) remains the leading cause of death in the western world. Despite advancements in interventional revascularization technologies, many patients are not candidates for them due to comorbidities or lack of local resources. Non-invasive approaches to accelerate revascularization within ischemic tissues through angiogenesis by providing Vascular Endothelial Growth Factor (VEGF) in protein or gene form has been effective in animal models but not in humans likely due to its short half-life and systemic toxicity. Here, we tested the hypothesis that PR1P, a small VEGF binding peptide that we developed, which stabilizes and upregulates endogenous VEGF, could be used to improve outcome from MI in rodents. To test this hypothesis, we induced MI in mice and rats via left coronary artery ligation and then treated animals with every other day intraperitoneal PR1P or scrambled peptide for 14 days. Hemodynamic monitoring and echocardiography in mice and echocardiography in rats at 14 days showed PR1P significantly improved multiple functional markers of heart function, including stroke volume and cardiac output. Furthermore, molecular biology and histological analyses of tissue samples showed that systemic PR1P targeted, stabilized and upregulated endogenous VEGF within ischemic myocardium. We conclude that PR1P is a potential non-invasive candidate therapeutic for MI.


Subject(s)
AC133 Antigen/metabolism , Disease Models, Animal , Ischemia/complications , Myocardial Infarction/prevention & control , Neovascularization, Physiologic/drug effects , Peptide Fragments/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Animals , Female , Ischemia/metabolism , Ischemia/pathology , Mice , Mice, Inbred C57BL , Myocardial Infarction/etiology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Rats , Rats, Sprague-Dawley , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Ventricular Function, Left/drug effects
5.
PLoS Genet ; 13(6): e1006848, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28617813

ABSTRACT

Recent findings indicate that growth factor-driven angiogenesis is markedly influenced by genetic variation. This variation in angiogenic responsiveness may alter the susceptibility to a number of angiogenesis-dependent diseases. Here, we utilized the genetic diversity available in common inbred mouse strains to identify the loci and candidate genes responsible for differences in angiogenic response. The corneal micropocket neovascularization assay was performed on 42 different inbred mouse strains using basic fibroblast growth factor (bFGF) pellets. We performed a genome-wide association study utilizing efficient mixed-model association (EMMA) mapping using the induced vessel area from all strains. Our analysis yielded five loci with genome-wide significance on chromosomes 4, 8, 11, 15 and 16. We further refined the mapping on chromosome 4 within a haplotype block containing multiple candidate genes. These genes were evaluated by expression analysis in corneas of various inbred strains and in vitro functional assays in human microvascular endothelial cells (HMVECs). Of these, we found the expression of peptidyl arginine deiminase type II (Padi2), known to be involved in metabolic pathways, to have a strong correlation with a haplotype shared by multiple high angiogenic strains. In addition, inhibition of Padi2 demonstrated a dosage-dependent effect in HMVECs. To investigate its role in vivo, we knocked down Padi2 in transgenic kdrl:zsGreen zebrafish embryos using morpholinos. These embryos had disrupted vessel formation compared to control siblings. The impaired vascular pattern was partially rescued by human PADI2 mRNA, providing evidence for the specificity of the morphant phenotype. Taken together, our study is the first to indicate the potential role of Padi2 as an angiogenesis-regulating gene. The characterization of Padi2 and other genes in associated pathways may provide new understanding of angiogenesis regulation and novel targets for diagnosis and treatment of a wide variety of angiogenesis-dependent diseases.


Subject(s)
Genome-Wide Association Study , Hydrolases/genetics , Neovascularization, Pathologic/genetics , Animals , Endothelial Cells/metabolism , Endothelial Cells/pathology , Fibroblast Growth Factor 2/genetics , Genetic Variation , Haplotypes , Humans , Hydrolases/biosynthesis , Mice , Mice, Inbred Strains , Phenotype , Protein-Arginine Deiminase Type 2 , Protein-Arginine Deiminases , Zebrafish
6.
Angiogenesis ; 20(3): 399-408, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28397127

ABSTRACT

Therapeutic angiogenesis is an experimental frontier in vascular biology that seeks to deliver angiogenic growth factors to ischemic or injured tissues to promote targeted formation of new blood vessels as an alternative approach to surgical revascularization procedures. Vascular endothelial growth factor (VEGF) is a potent angiogenic signal protein that is locally upregulated at sites of tissue injury. However, therapies aimed at increasing VEGF levels experimentally by injecting VEGF gene or protein failed to improve outcomes in human trials in part due to its short half-life and systemic toxicity. We recently designed a novel 12-amino acid peptide (PR1P) whose sequence was derived from an extracellular VEGF-binding domain of the pro-angiogenic glycoprotein prominin-1. In this study, we characterized the molecular binding properties of this novel potential therapeutic for targeted angiogenesis and provided the foundation for its use as an angiogenic molecule that can potentiate endogenous VEGF. We showed that PR1P bound VEGF directly and enhanced VEGF binding to endothelial cells and to VEGF receptors VEGFR2 and neuropilin-1. PR1P increased angiogenesis in the murine corneal micropocket assay when combined with VEGF, but had no activity without added VEGF. In addition, PR1P also enhanced angiogenesis in murine choroidal neovascularization and wound-healing models and augmented reperfusion in a murine hind-limb ischemia model. Together our data suggest that PR1P enhanced angiogenesis by potentiating the activity of endogenous VEGF. In so doing, this novel therapy takes advantage of endogenous VEGF gradients generated in injured tissues and may improve the efficacy of and avoid systemic toxicity seen with previous VEGF therapies.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Neovascularization, Physiologic/drug effects , Peptide Fragments/pharmacology , Peptides/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Animals , Female , Humans , Ischemia/pathology , Mice, Inbred C57BL , Perfusion , Protein Binding/drug effects
7.
FASEB J ; 29(2): 662-70, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25406462

ABSTRACT

The incidence of certain angiogenesis-dependent diseases is higher in Caucasians than in African Americans. Angiogenesis is amplified in wound healing and cornea models in albino C57 mice compared with black C57 mice. Moreover, mouse and human melanocytes with low pigmentation stimulate endothelial cell (EC) proliferation and migration in vitro more than melanocytes with high pigmentation. This effect is due, in part, to the secretion of an angiogenic protein called fibromodulin (FMOD) from lowly pigmented melanocytes. Herein, we expand upon the mechanism contributing to increased angiogenesis in lighter skin and report that monocyte chemotactic protein-1 (MCP-1) is secreted by nonpigmented mouse melanocytes by 5- to 10-fold more than pigmented melanocytes. MCP-1 protein stimulates EC proliferation and migration in vitro and angiogenesis in vivo. Mechanistic studies determine that FMOD is upstream of MCP-1 and promotes its secretion from both melanocytes and activated ECs via stimulation of NF-κB activity. Mice injected with FMOD-neutralizing antibodies show 2.3-fold decreased levels of circulating MCP-1. Human studies confirmed that, on average, Caucasians have 2-fold higher serum levels of MCP-1 than African Americans. Taken together, this study implicates the FMOD/MCP-1 pathway in the regulation of angiogenesis by local melanocytes and suggests that melanogenic activity may protect against aberrant angiogenic diseases.


Subject(s)
Chemokine CCL2/metabolism , Melanocytes/cytology , Neovascularization, Pathologic , Skin Pigmentation , Black or African American , Angiogenesis Inducing Agents/metabolism , Animals , Cells, Cultured , Endothelial Cells/cytology , Enzyme-Linked Immunosorbent Assay , Extracellular Matrix Proteins/metabolism , Fibromodulin , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microcirculation , NF-kappa B/metabolism , Pigmentation , Proteoglycans/metabolism
8.
Nanomedicine ; 11(8): 1993-2002, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26247432

ABSTRACT

Oral delivery of poorly soluble and permeable drugs represents a significant challenge in drug development. The oral delivery of drugs remains to be the ultimate route of any drugs. However, in many cases, drugs are not absorbed well in the gastrointestinal tract, or they lose their activity. Polymer micelles were recognized as an effective carrier system for drug encapsulation, and are now studied as a vehicle for oral delivery of insoluble compounds. We characterized the properties of monomethoxy polyethylene glycol-poly lactic acid (mPEG-PLA) micelles, and visualized their internalization in mouse small intestine. Using Caco-2 cells as a cellular model, we studied the kinetics of particle uptake, their transport, and the molecular mechanism of their intestinal absorption. Moreover, by inhibiting specific endocytosis pathways, pharmacologically and genetically, we found that mPEG-PLA nanoparticle endocytosis is mediated by clathrin in an energy-dependent manner, and that the low-density lipoprotein receptor is involved. FROM THE CLINICAL EDITOR: Many current drugs used are non-water soluble and indeed, the ability to deliver these drugs via the gastrointestinal tract remains the holy grail for many researchers. The authors in this paper developed monomethoxy polyethylene glycol-poly lactic acid (mPEG-PLA) micelles as a drug nanocarrier, and studied the mechanism of uptake across intestinal cells. The findings should improve our current understanding and point to the development of more nanocarriers.


Subject(s)
Drug Carriers/pharmacokinetics , Intestine, Small/metabolism , Lactic Acid/pharmacokinetics , Micelles , Polyethylene Glycols/pharmacokinetics , Polymers/pharmacokinetics , Administration, Oral , Animals , Caco-2 Cells , Drug Carriers/chemistry , Endocytosis , Humans , Intestinal Absorption , Lactic Acid/chemistry , Mice, Inbred C57BL , Polyesters , Polyethylene Glycols/chemistry , Polymers/chemistry
9.
Angiogenesis ; 16(3): 639-46, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23423728

ABSTRACT

We have observed substantial differences in angiogenic responsiveness in mice and have mapped the genetic loci responsible for these differences. We have found that the albino mutation is one of the loci responsible for such differences. Using B6.A consomic strains, we determined that chromosome 7 bears a locus that inhibits VEGF-induced corneal neovascularization. F2 crosses between B6.A consomic mice and C57BL/6J parents along with AXB and BXA recombinant inbred strains demonstrated highest linkage near the tyrosinase gene. This region was named AngVq4. Congenic animals confirmed this locus, but could not demonstrate that the classical tyrosinase albino (c) mutation was causative because of the existence of additional linked loci in the congenic region. However, in 1970, a second tyrosinase albino mutation (c-2J) arose in the C57BL/6J background at Jackson Labs. Testing this strain (C57BL/6J) demonstrated that the albino mutation is sufficient to completely explain the alteration in angiogenic response that we observed in congenic animals. Thus, we conclude that the classical tyrosinase mutation is responsible for AngVq4. In contrast to the cornea, where pigmented animals exhibit increased angiogenic responsiveness, iris neovascularization was inhibited in pigmented animals. These results may partially explain increased aggressiveness in amelanotic melanoma, as well as ethnic differences in diabetic retinopathy and macular degeneration.


Subject(s)
Albinism, Oculocutaneous/genetics , Corneal Neovascularization/enzymology , Monophenol Monooxygenase/genetics , Mutation/genetics , Animals , Corneal Neovascularization/genetics , Crosses, Genetic , Endothelial Cells/physiology , Genetic Linkage , Genotype , Humans , Mice , Mice, Inbred Strains
10.
Angiogenesis ; 16(2): 405-16, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23150059

ABSTRACT

Prominin-1, a pentaspan transmembrane protein, is a unique cell surface marker commonly used to identify stem cells, including endothelial progenitor cells and cancer stem cells. However, recent studies have shown that prominin-1 expression is not restricted to stem cells but also occurs in modified forms in many mature adult human cells. Although prominin-1 has been studied extensively as a stem cell marker, its physiological function of the protein has not been elucidated. We investigated prominin-1 function in two cell lines, primary human endothelial cells and B16-F10 melanoma cells, both of which express high levels of prominin-1. We found that prominin-1 directly interacts with the angiogenic and tumor survival factor vascular endothelial growth factor (VEGF) in both the primary endothelial cells and the melanoma cells. Knocking down prominin-1 in the endothelial cells disrupted capillary formation in vitro and decreased angiogenesis in vivo. Similarly, tumors derived from prominin-1 knockdown melanoma cells had a reduced growth rate in vivo. Further, melanoma cells with knocked down prominin-1 had diminished ability to interact with VEGF, which was associated with decreased bcl-2 protein levels and increased apoptosis. In vitro studies with soluble prominin-1 showed that it stabilized dimer formation of VEGF164, but not VEGF121. Taken together, our findings support the notion that prominin-1 plays an active role in cell growth through its ability to interact and potentiate the anti-apoptotic and pro-angiogenic activities of VEGF. Additionally, prominin-1 promotes tumor growth by supporting angiogenesis and inhibiting tumor cell apoptosis.


Subject(s)
Antigens, CD/metabolism , Glycoproteins/metabolism , Peptides/metabolism , Vascular Endothelial Growth Factor A/metabolism , AC133 Antigen , Apoptosis , Blotting, Western , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Melanoma/pathology , Protein Binding , Real-Time Polymerase Chain Reaction
11.
Angiogenesis ; 16(3): 541-51, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23334337

ABSTRACT

Endometriosis is an angiogenesis-dependent disease. Many studies demonstrated inhibition of angiogenesis leads to inhibition of endometriotic growth, however underlying mechanism is still not fully understood. Our previous study suggested vascular endothelial growth factor C (VEGF-C) as a target of anti-angiogenesis therapy for endometriosis. In this study, VEGF-C in endometrium and its role in angiogenesis of endometriosis were studied. Human endometrium were obtained from women with and without endometriosis for molecular studies. VEGF-A, VEGF-B, VEGF-C and VEGF-D mRNA and proteins in eutopic and ectopic endometrium were measured. Human endothelial cells were transfected with VEGF-C siRNA in vitro, effects of VEGF-C on endothelial cell migration, invasion and tube formation were investigated in vitro. Angiogenesis was inhibited in wild type mice, vascular permeability in dermal skin was determined in vivo. Transplanted endometrium were inhibited by VEGF-C siRNA in immunocompromised mice, development, growth and angiogenesis of the experimental endometriosis were compared in vivo. The results showed that VEGF-C mRNA and protein were increased in eutopic and ectopic endometrium of endometriosis patients. VEGF-C siRNA significantly inhibited endothelial cell migration and tube formation. VEGF-C siRNA significantly inhibited development and angiogenesis of the experimental endometriotic lesions in mice. Supplementation and over-expression of VEGF-C significantly reversed the inhibitory effects on the endothelial functions, vascular permeability and endometriotic growth. In conclusion, VEGF-C is increased in endometrium and it promotes endothelial functions, vascular permeability and development of experimental endometriosis. VEGF-C is important for angiogenesis in endometriosis.


Subject(s)
Capillary Permeability/physiology , Endometriosis/metabolism , Endometrium/metabolism , Endothelial Cells/physiology , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor C/metabolism , Analysis of Variance , Animals , Cell Movement/drug effects , Cell Movement/physiology , Endometrium/cytology , Endothelial Cells/metabolism , Female , Humans , Mice , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology
12.
Cancer Invest ; 31(3): 177-82, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23402284

ABSTRACT

INTRODUCTION: This study evaluated objective response and safety for the combination of granulocyte macrophage-colony stimulating factor (GM-CSF), ketoconazole, and mitoxantrone in castration-resistant prostate cancer (CRPC) patients who previously failed docetaxel-based chemotherapy. METHODS: Treatment consisted of 400 mg TID ketoconazole, 12 mg/m(2) mitoxantrone every 3 weeks, and 250 µg/m(2) GM-CSF. RESULTS: Twenty-nine patients were evaluable for response. Median overall survival (OS) for all patients was 18.03 months. Patients with a higher PSA decrease experienced an increased OS and progression-free survival (PFS). CONCLUSION: This combination demonstrated significant antitumor activity with reversible toxicity in CRPC patients who previously failed docetaxel-based therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Prostatic Neoplasms/drug therapy , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Docetaxel , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Humans , Ketoconazole/administration & dosage , Male , Middle Aged , Mitoxantrone/administration & dosage , Orchiectomy , Prostate-Specific Antigen/blood , Prostatic Neoplasms/blood , Prostatic Neoplasms/mortality , Taxoids/administration & dosage
13.
Cancer Cell ; 7(1): 101-11, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15652753

ABSTRACT

Development of antiangiogenic therapies would be significantly facilitated by quantitative surrogate pharmacodynamic markers. Circulating peripheral blood endothelial cells (CECs) and/or their putative progenitor subset (CEPs) have been proposed but not yet fully validated for this purpose. Herein, we provide such validation by showing a striking correlation between highly genetically heterogeneous bFGF- or VEGF-induced angiogenesis and intrinsic CEC or CEP levels measured by flow cytometry, among eight different inbred mouse strains. Moreover, studies using genetically altered mice showed that levels of these cells are affected by regulators of angiogenesis, including VEGF, Tie-2, and thrombospondin-1. Finally, treatment with a targeted VEGFR-2 antibody caused a dose-dependent reduction in viable CEPs that precisely paralleled its previously and empirically determined antitumor activity.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Biomarkers , Endothelial Cells/physiology , Neovascularization, Pathologic , Stem Cells/physiology , Angiogenesis Inhibitors/metabolism , Animals , Biological Assay/methods , Cell Survival , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fibroblast Growth Factors/metabolism , Flow Cytometry , Humans , Male , Mice , Mice, Inbred Strains , Mice, Transgenic , Phenotype , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Stem Cells/drug effects , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
14.
Angiogenesis ; 15(2): 265-73, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22402885

ABSTRACT

Platelets sequester angiogenesis regulatory proteins which suggests an avenue for developing biomarkers to monitor disease. We describe a comparison of angiogenesis regulatory proteins found in platelets of colorectal cancer patients and normal controls. Platelet and plasma content of vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), platelet derived growth factor (PDGF), platelet factor 4 (PF4), thrombospondin-1 (TSP-1) and endostatin in 35 patients with colon cancer were compared with 84 age-matched healthy controls using ELISAs. We standardized the platelet preparation procedure, introduced process controls and normalized the respective protein levels to platelet numbers using an actin ELISA. Statistically significant differences were found in the median levels of VEGF, PF4 and PDGF in platelets of patients with cancer compared to healthy individuals. Platelet concentrations in cancer patients versus controls were: VEGF 1.3 versus 0.6 pg/10(6), PF4 18.5 versus 9.4 ng/10(6), and PDGF 34.1 versus 21.0 pg/10(6). Multivariable logistic regression analysis indicated that PDGF, PF4 and VEGF were independent predictors of colorectal carcinoma and as a set provided statistically significant discrimination (area under the curve = 0.893, P < .0001). No significant differences were detected for bFGF, endostatin, or TSP-1. Reference Change Value analysis determined that the differences seen were not clinically significant. Plasma levels yielded no correlations.


Subject(s)
Colorectal Neoplasms/blood , Neovascularization, Pathologic/blood , Platelet Factor 4/blood , Platelet-Derived Growth Factor/metabolism , Vascular Endothelial Growth Factor A/blood , Adult , Aged , Colorectal Neoplasms/pathology , Female , Fibroblast Growth Factor 2/blood , Humans , Male , Middle Aged , Neovascularization, Pathologic/pathology , Thrombospondin 1/blood
15.
Am J Pathol ; 178(4): 1782-91, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21435458

ABSTRACT

Endometriosis is a debilitating disease characterized by the growth of ectopic endometrial tissue. It is widely accepted that angiogenesis plays an integral part in the establishment and growth of endometriotic lesions. Recent data from a variety of angiogenesis-dependent diseases suggest a critical role of bone marrow-derived endothelial progenitor cells (EPCs) in neovascularization. In this study we examined the blood levels of EPCs and mature circulating endothelial cells in a mouse model of surgically induced endometriosis. Fluorescence-activated cell sorting analysis revealed elevated levels of EPCs in the blood of mice with endometriosis compared with control subject that underwent a sham operation. EPC concentrations positively correlated with the amount of endometriotic tissue and peaked 1 to 4 days after induction of disease. In a green fluorescent protein bone marrow transplant experiment we found green fluorescent protein-positive endothelial cells incorporated into endometriotic lesions but not eutopic endometrium, as revealed by flow cytometry and immunohistochemistry. Finally, treatment of endometriosis-bearing mice with the angiogenesis inhibitor Lodamin, an oral nontoxic formulation of TNP-470, significantly decreased EPC levels while suppressing lesion growth. Taken together, our data indicate an important role for bone marrow-derived endothelial cells in the pathogenesis of endometriosis and support the potential clinical use of anti-angiogenic therapy as a novel treatment modality for this disease.


Subject(s)
Endometriosis/blood , Endothelial Cells/cytology , Stem Cells/cytology , Administration, Oral , Angiogenesis Inhibitors/pharmacology , Animals , Bone Marrow Transplantation , Cell Separation , Cyclohexanes/pharmacology , Disease Models, Animal , Endometriosis/pathology , Female , Flow Cytometry , Green Fluorescent Proteins/metabolism , Immunohistochemistry/methods , Mice , Mice, Inbred C57BL , O-(Chloroacetylcarbamoyl)fumagillol , Sesquiterpenes/pharmacology , Up-Regulation
16.
Ann Surg Oncol ; 19(5): 1685-91, 2012 May.
Article in English | MEDLINE | ID: mdl-22130619

ABSTRACT

BACKGROUND: Our aim was to evaluate the changes in age, stage distribution, and overall survival (OS) of patients with esophageal adenocarcinoma (EAC) over time. METHODS: Patients from the Surveillance, Epidemiology, and End Results (SEER) database aged ≥ 20 with invasive EAC, diagnosed from 1973-2003 were reviewed. Survival follow-up ended in 2006. RESULTS: There were 11,620 patients; 6580 (57%) aged ≥ 65. The stage distribution was 22%, 35%, and 43% for localized, regional, and distant metastasis for patients aged <65, and 33%, 33%, and 34% for patients aged ≥ 65. The number of patients ≥ 65 years with localized stage increased over time. Three-year OS for localized, regional, and distant disease increased from 19%, 10%, and 1% in 1973-1976, to 34%, 13%, and 2% in 1987-1991, and to 45%, 25%, and 4% in 2002-2003 (P < 0.001). A sub-analysis of 5475 patients from 1988-2002 showed better survival for patients with esophagectomy for all stages. Three-year OS for 2074 patients with esophagectomy improved every 5 years from 1988-2002 (39%, 43% to 54%, P < 0.001). Stratified by stage, year and esophagectomy status, patients aged <65 had better survival compared to patients aged ≥ 65 (P < 0.001). CONCLUSIONS: There has been a substantial improvement in overall survival among patients with invasive EAC over the last 3 decades. Patients receiving esophagectomy had longer survival. Survival with esophagectomy improved in each time period. Although younger EAC patients were diagnosed at more advanced stages over time, they had better survival.


Subject(s)
Adenocarcinoma/mortality , Adenocarcinoma/pathology , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Adenocarcinoma/epidemiology , Adenocarcinoma/surgery , Adult , Age Distribution , Age Factors , Aged , Cohort Studies , Esophageal Neoplasms/epidemiology , Esophageal Neoplasms/surgery , Esophagectomy , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Staging , Population Surveillance , Retrospective Studies , Survival Rate , United States/epidemiology , Young Adult
17.
Cancer Invest ; 30(5): 415-21, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22571342

ABSTRACT

BACKGROUND: In carcinogenesis, methylation of DNA promoter regions results in inactivation of tumor-suppressing genes. MG98 was designed to inhibit DNA methyltransferases enzyme 1 production. METHODS: This multicenter study explored two schedules of MG98 with Interferon-α-2ß to identify schedule and dose for patients with metastatic RCC. RESULTS: Doses of IFN 9 MIU/MG98 125 mg/m(2) for a continuous schedule and IFN 9 MIU/MG98 200 mg/m(2) for an intermittent schedule were considered the MTDs. Treatment resulted in one PR and eight SD. CONCLUSION: MG98 combined with IFN was safe and resulted in clinical activity.


Subject(s)
Carcinoma, Renal Cell/drug therapy , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , Kidney Neoplasms/drug therapy , Oligodeoxyribonucleotides/therapeutic use , Thionucleotides/therapeutic use , Adult , Aged , Carcinoma, Renal Cell/mortality , DNA (Cytosine-5-)-Methyltransferase 1 , Disease-Free Survival , Humans , Kidney Neoplasms/mortality , Middle Aged , Oligodeoxyribonucleotides/adverse effects , Thionucleotides/adverse effects
18.
Acta Oncol ; 51(5): 584-8, 2012 May.
Article in English | MEDLINE | ID: mdl-22248089

ABSTRACT

BACKGROUND: To investigate the utility of stereotactic body radiotherapy (SBRT) in the treatment of painful renal cell carcinoma (RCC) bone metastases, and for a possible dose effect on time to symptom relief. MATERIAL AND METHODS: Eighteen patients with 24 painful osseous lesions from metastatic RCC were treated with SBRT. The most common treatment regimens were 24 Gy in 3 fractions and 40 Gy in 5 fractions. The times from treatment to first reported pain relief and time to symptom recurrence were evaluated. Median follow-up was 38 weeks (1-156 weeks). RESULTS: Seventy-eight percent of all patients had pain relief. Patients treated with a BED > 85 Gy achieved faster and more durable pain relief compared to those treated with a BED < 85 Gy. There was decrease in time to pain relief after a change in treatment regimen to 8 Gy × 5 fractions (BED = 86). There was only one patient with grade 1 skin toxicity. No neurological or other toxicity was observed. CONCLUSIONS: SBRT can safely and effectively treat painful RCC bony metastases. There appears to be a relationship between radiation dose and time to stable pain relief.


Subject(s)
Bone Neoplasms/surgery , Carcinoma, Renal Cell/surgery , Kidney Neoplasms/surgery , Neoplasm Recurrence, Local/surgery , Pain/etiology , Pain/prevention & control , Radiosurgery , Bone Neoplasms/secondary , Carcinoma, Renal Cell/pathology , Dose Fractionation, Radiation , Follow-Up Studies , Humans , Kidney Neoplasms/pathology , Neoplasm Recurrence, Local/pathology , Prognosis , Survival Rate , Time Factors
19.
Future Oncol ; 8(3): 231-7, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22409460

ABSTRACT

This paper reviews the development of the combination of modified vaccinia Ankara (MVA) to deliver the tumor-associated antigen 5T4 as a novel immunotherapeutic vaccine. The oncofetal antigen 5T4 is highly expressed in 80% of breast, kidney, colorectal, prostate and ovarian carcinomas, making it an ideal antigen for vaccine therapy. To date, more than 3000 doses of MVA-5T4 have been administered to colorectal, renal and prostate cancer patients, with rare occurrences of grade 3 or 4 vaccination-related adverse events being observed. Studies have demonstrated that MVA-5T4 is safe and highly immunogenic, both as monotherapy and in combination with other standard of care therapies. Although an immune response has been observed, antitumor activity has been modest or absent in clinical trials. A Phase III trial resulted in the development of an immune response surrogate that is to be applied to all future MVA-5T4 clinical trials. With minimal side effects and the ability to produce a strong immunogenic response, MVA-5T4 is a viable addition to the cancer therapy arsenal.


Subject(s)
Colorectal Neoplasms/drug therapy , Immunotherapy/methods , Kidney Neoplasms/drug therapy , Membrane Glycoproteins/immunology , Prostatic Neoplasms/drug therapy , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Clinical Trials as Topic , Colorectal Neoplasms/immunology , Colorectal Neoplasms/prevention & control , Drug Delivery Systems , Genetic Vectors , Humans , Kidney Neoplasms/immunology , Kidney Neoplasms/prevention & control , Male , Membrane Glycoproteins/pharmacology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/prevention & control , Vaccination/adverse effects , Vaccination/methods , Vaccinia virus/immunology
20.
J Clin Invest ; 132(23)2022 12 01.
Article in English | MEDLINE | ID: mdl-36453543

ABSTRACT

Glaucoma is a highly heritable disease that is a leading cause of blindness worldwide. Here, we identified heterozygous thrombospondin 1 (THBS1) missense alleles altering p.Arg1034, a highly evolutionarily conserved amino acid, in 3 unrelated and ethnically diverse families affected by congenital glaucoma, a severe form of glaucoma affecting children. Thbs1R1034C-mutant mice had elevated intraocular pressure (IOP), reduced ocular fluid outflow, and retinal ganglion cell loss. Histology revealed an abundant, abnormal extracellular accumulation of THBS1 with abnormal morphology of juxtacanalicular trabecular meshwork (TM), an ocular tissue critical for aqueous fluid outflow. Functional characterization showed that the THBS1 missense alleles found in affected individuals destabilized the THBS1 C-terminus, causing protein misfolding and extracellular aggregation. Analysis using a range of amino acid substitutions at position R1034 showed that the extent of aggregation was correlated with the change in protein-folding free energy caused by variations in amino acid structure. Extracellular matrix (ECM) proteins, especially fibronectin, which bind to THBS1, also accumulated within THBS1 deposits. These results show that missense variants altering THBS1 p.Arg1034 can cause elevated IOP through a mechanism involving impaired TM fluid outflow in association with accumulation of aggregated THBS1 in the ECM of juxtacanalicular meshwork with altered morphology.


Subject(s)
Glaucoma , Trabecular Meshwork , Animals , Mice , Extracellular Matrix Proteins , Thrombospondin 1/genetics , Alleles , Glaucoma/genetics , Amino Acids
SELECTION OF CITATIONS
SEARCH DETAIL