Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Int J Mol Sci ; 24(19)2023 Sep 22.
Article in English | MEDLINE | ID: mdl-37833857

ABSTRACT

The impairment in microvascular network formation could delay the restoration of blood flow after acute limb ischemia. A high-content screen of a GSK-published kinase inhibitor library identified a set of ROCK inhibitor hits enhancing endothelial network formation. Subsequent kinase activity profiling against a panel of 224 protein kinases showed that two indazole-based ROCK inhibitor hits exhibited high selectivity for ROCK1 and ROCK2 isoforms compared to other ROCK inhibitors. One of the chemical entities, GSK429286, was selected for follow-up studies. We found that GSK429286 was ten times more potent in enhancing endothelial tube formation than Fasudil, a classic ROCK inhibitor. ROCK1 inhibition by RNAi phenocopied the angiogenic phenotype of the GSK429286 compound. Using an organotypic angiogenesis co-culture assay, we showed that GSK429286 formed a dense vascular network with thicker endothelial tubes. Next, mice received either vehicle or GSK429286 (10 mg/kg i.p.) for seven days after hindlimb ischemia induction. As assessed by laser speckle contrast imaging, GSK429286 potentiated blood flow recovery after ischemia induction. At the histological level, we found that GSK429286 significantly increased the size of new microvessels in the regenerating areas of ischemic muscles compared with vehicle-treated ones. Our findings reveal that selective ROCK inhibitors have in vitro pro-angiogenic properties and therapeutic potential to restore blood flow in limb ischemia.


Subject(s)
Endothelial Cells , rho-Associated Kinases , Mice , Animals , rho-Associated Kinases/metabolism , Endothelial Cells/metabolism , Neovascularization, Physiologic , Ischemia/metabolism , Regional Blood Flow , Hindlimb/pathology , Disease Models, Animal , Muscle, Skeletal/metabolism
2.
Mol Genet Genomic Med ; 11(8): e2178, 2023 08.
Article in English | MEDLINE | ID: mdl-37147786

ABSTRACT

BACKGROUND: Dyslipidemia is recognized as one of the risk factors of cardiovascular diseases (CVDs), type 2 diabetes mellitus (T2DM), and non-alcoholic fatty liver disease (NAFLD). OBJECTIVE: The study aimed to investigate the association between selected single nucleotide polymorphisms (SNPs) with dyslipidemia and increased susceptibility risks of CVD, NAFLD, and/or T2DM in dyslipidemia patients in comparison with healthy control individuals from the Qatar genome project. METHODS: A community-based cross-sectional study was conducted among 2933 adults (859 dyslipidemia patients and 2074 healthy control individuals) from April to December 2021 to investigate the association between 331 selected SNPs with dyslipidemia and increased susceptibility risks of CVD, NAFLD and/or T2DM, and covariates. RESULTS: The genotypic frequencies of six SNPs were found to be significantly different in dyslipidemia patients subjects compared to the control group among males and females. In males, three SNPs were found to be significant, the rs11172113 in over-dominant model, the rs646776 in recessive and over-dominant models, and the rs1111875 in dominant model. On the other hand, two SNPs were found to be significant in females, including rs2954029 in recessive model, and rs1801251 in dominant and recessive models. The rs17514846 SNP was found for dominant and over-dominant models among males and only the dominant model for females. We found that the six SNPs linked to gender type had an influence in relation to disease susceptibility. When controlling for the four covariates (gender, obesity, hypertension, and diabetes), the difference between dyslipidemia and the control group remained significant for the six variants. Finally, males were three times more likely to have dyslipidemia in comparison with females, hypertension was two times more likely to be present in the dyslipidemia group, and diabetes was six times more likely to be in the dyslipidemia group. CONCLUSION: The current investigation provides evidence of association for a common SNP to coronary heart disease and suggests a sex-dependent effect and encourage potential therapeutic applications.


Subject(s)
Cardiovascular Diseases , Diabetes Mellitus, Type 2 , Dyslipidemias , Hypertension , Non-alcoholic Fatty Liver Disease , Adult , Male , Female , Humans , Polymorphism, Single Nucleotide , Diabetes Mellitus, Type 2/genetics , Qatar/epidemiology , Cross-Sectional Studies , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/genetics , Cardiovascular Diseases/complications , Dyslipidemias/epidemiology , Dyslipidemias/genetics , Dyslipidemias/complications
3.
Front Oncol ; 13: 1269376, 2023.
Article in English | MEDLINE | ID: mdl-38239643

ABSTRACT

Metastasis is the cause of most triple-negative breast cancer deaths, yet anti-metastatic therapeutics remain limited. To develop new therapeutics to prevent metastasis, pathophysiologically relevant assays that recapitulate tumor microenvironment is essential for disease modeling and drug discovery. Here, we have developed a microfluidic metastasis-on-chip assay of the early stages of cancer metastasis integrated with the triple-negative breast cancer cell line (MDA-MB-231), stromal fibroblasts and a perfused microvessel. High-content imaging with automated quantification methods was optimized to assess the tumor cell invasion and intravasation within the model. Cell invasion and intravasation were enhanced when fibroblasts co-cultured with a breast cancer cell line (MDA-MB-231). However, the non-invasive breast cancer cell line, MCF7, remained non-invasive in our model, even in the presence of fibroblasts. High-content screening of a targeted anti-cancer therapy drug library was conducted to evaluate the drug response sensitivity of the optimized model. Through this screening, we identified 30 compounds that reduced the tumor intravasation by 60% compared to controls. Multi-parametric phenotypic analysis was applied by combining the data from the metastasis-on-chip, cell proliferation and 2D cell migration screens, revealing that the drug library was clustered into eight distinct groups with similar drug responses. Notably, MEK inhibitors were enriched in cluster cell invasion and intravasation. In contrast, drugs with molecular targets: ABL, KIT, PDGF, SRC, and VEGFR were enriched in the drug clusters showing a strong effect on tumor cell intravasation with less impact on cell invasion or cell proliferation, of which, Imatinib, a multi-kinase inhibitor targeting BCR-ABL/PDGFR/KIT. Further experimental analysis showed that Imatinib enhanced endothelial barrier stability as measured by trans-endothelial electrical resistance and significantly reduced the trans-endothelial invasion activity of tumor cells. Our findings demonstrate the potential of our metastasis-on-chip assay as a powerful tool for studying cancer metastasis biology, drug discovery aims, and assessing drug responses, offering prospects for personalized anti-metastatic therapies for triple-negative breast cancer patients.

4.
Int J Mol Sci ; 23(9)2022 Apr 28.
Article in English | MEDLINE | ID: mdl-35563280

ABSTRACT

Angiogenesis is a critical cellular process toward establishing a functional circulatory system capable of delivering oxygen and nutrients to the tissue in demand. In vitro angiogenesis assays represent an important tool for elucidating the biology of blood vessel formation and for drug discovery applications. Herein, we developed a novel, high content 2D angiogenesis assay that captures endothelial morphogenesis's cellular processes, including lumen formation. In this assay, endothelial cells form luminized vascular-like structures in 48 h. The assay was validated for its specificity and performance. Using the optimized assay, we conducted a phenotypic screen of a library containing 150 FDA-approved cardiovascular drugs to identify modulators of lumen formation. The screening resulted in several L-type calcium channel blockers being able to expand the lumen space compared to controls. Among these blockers, Lacidipine was selected for follow-up studies. We found that the endothelial cells treated with Lacidipine showed enhanced activity of caspase-3 in the luminal space. Pharmacological inhibition of caspase activity abolished the Lacidipine-enhancing effect on lumen formation, suggesting the involvement of apoptosis. Using a Ca2+ biosensor, we found that Lacipidine reduces the intracellular Ca2+ oscillations amplitude in the endothelial cells at the early stage, whereas Lacidipine blocks these Ca2+ oscillations completely at the late stage. The inhibition of MLCK exhibits a phenotype of lumen expansion similar to that of Lacidipine. In conclusion, this study describes a novel high-throughput phenotypic assay to study angiogenesis. Our findings suggest that calcium signalling plays an essential role during lumen morphogenesis. L-type Ca2+ channel blockers could be used for more efficient angiogenesis-mediated therapies.


Subject(s)
Calcium Channel Blockers , Dihydropyridines , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Dihydropyridines/pharmacology , Dihydropyridines/therapeutic use , Endothelial Cells , Morphogenesis
5.
Int J Mol Sci ; 22(19)2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34638923

ABSTRACT

Arteriogenesis is one of the primary physiological means by which the circulatory collateral system restores blood flow after significant arterial occlusion in peripheral arterial disease patients. Vascular smooth muscle cells (VSMCs) are the predominant cell type in collateral arteries and respond to altered blood flow and inflammatory conditions after an arterial occlusion by switching their phenotype between quiescent contractile and proliferative synthetic states. Maintaining the contractile state of VSMC is required for collateral vascular function to regulate blood vessel tone and blood flow during arteriogenesis, whereas synthetic SMCs are crucial in the growth and remodeling of the collateral media layer to establish more stable conduit arteries. Timely VSMC phenotype switching requires a set of coordinated actions of molecular and cellular mediators to result in an expansive remodeling of collaterals that restores the blood flow effectively into downstream ischemic tissues. This review overviews the role of VSMC phenotypic switching in the physiological arteriogenesis process and how the VSMC phenotype is affected by the primary triggers of arteriogenesis such as blood flow hemodynamic forces and inflammation. Better understanding the role of VSMC phenotype switching during arteriogenesis can identify novel therapeutic strategies to enhance revascularization in peripheral arterial disease.


Subject(s)
Arteries/physiology , Cell Proliferation/physiology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/physiology , Vascular Remodeling/physiology , Animals , Arterial Occlusive Diseases/genetics , Arterial Occlusive Diseases/metabolism , Arterial Occlusive Diseases/physiopathology , Arteries/cytology , Arteries/metabolism , Cell Proliferation/genetics , Collateral Circulation/genetics , Collateral Circulation/physiology , Gene Expression , Humans , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Phenotype , Vascular Remodeling/genetics
6.
Sci Rep ; 10(1): 13829, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32796870

ABSTRACT

Angiogenesis plays a key role in several diseases including cancer, ischemic vascular disease, and Alzheimer's disease. Chemical genetic screening of endothelial tube formation provides a robust approach for identifying signalling components that impact microvascular network morphology as well as endothelial cell biology. However, the analysis of the resulting imaging datasets has been limited to a few phenotypic features such as the total tube length or the number of branching points. Here we developed a high content analysis framework for detailed quantification of various aspects of network morphology including network complexity, symmetry and topology. By applying our approach to a high content screen of 1,280 characterised drugs, we found that drugs that result in a similar phenotype share the same mechanism of action or common downstream signalling pathways. Our multiparametric analysis revealed that a group of glutamate receptor antagonists enhances branching and network connectivity. Using an integrative meta-analysis approach, we validated the link between these receptors and angiogenesis. We further found that the expression of these genes is associated with the prognosis of Alzheimer's patients. In conclusion, our work shows that detailed image analysis of complex endothelial phenotypes can reveal new insights into biological mechanisms modulating the morphogenesis of endothelial networks and identify potential therapeutics for angiogenesis-related diseases.


Subject(s)
Endothelial Cells/pathology , Morphogenesis , Neovascularization, Pathologic/genetics , Receptors, Glutamate/physiology , Alzheimer Disease/etiology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Cells, Cultured , Humans , Signal Transduction
7.
Cardiovasc Res ; 116(11): 1863-1874, 2020 09 01.
Article in English | MEDLINE | ID: mdl-31584065

ABSTRACT

AIMS: Genome-wide association studies (GWAS) have consistently identified an association between coronary artery disease (CAD) and a locus on chromosome 10 containing a single gene, JCAD (formerly KIAA1462). However, little is known about the mechanism by which JCAD could influence the development of atherosclerosis. METHODS AND RESULTS: Vascular function was quantified in subjects with CAD by flow-mediated dilatation (FMD) and vasorelaxation responses in isolated blood vessel segments. The JCAD risk allele identified by GWAS was associated with reduced FMD and reduced endothelial-dependent relaxations. To study the impact of loss of Jcad on atherosclerosis, Jcad-/- mice were crossed to an ApoE-/- background and fed a high-fat diet from 6 to16 weeks of age. Loss of Jcad did not affect blood pressure or heart rate. However, Jcad-/-ApoE-/- mice developed significantly less atherosclerosis in the aortic root and the inner curvature of the aortic arch. En face analysis revealed a striking reduction in pro-inflammatory adhesion molecules at sites of disturbed flow on the endothelial cell layer of Jcad-/- mice. Loss of Jcad lead to a reduced recovery perfusion in response to hind limb ischaemia, a model of altered in vivo flow. Knock down of JCAD using siRNA in primary human aortic endothelial cells significantly reduced the response to acute onset of flow, as evidenced by reduced phosphorylation of NF-КB, eNOS, and Akt. CONCLUSION: The novel CAD gene JCAD promotes atherosclerotic plaque formation via a role in the endothelial cell shear stress mechanotransduction pathway.


Subject(s)
Aortic Diseases/metabolism , Atherosclerosis/metabolism , Cell Adhesion Molecules/metabolism , Coronary Artery Disease/metabolism , Coronary Circulation , Endothelium, Vascular/metabolism , Hindlimb/blood supply , Mechanotransduction, Cellular , Animals , Aorta/metabolism , Aorta/physiopathology , Aortic Diseases/genetics , Aortic Diseases/physiopathology , Aortic Diseases/prevention & control , Atherosclerosis/genetics , Atherosclerosis/physiopathology , Atherosclerosis/prevention & control , Cell Adhesion Molecules/genetics , Cells, Cultured , Coronary Artery Disease/genetics , Coronary Artery Disease/physiopathology , Coronary Vessels/metabolism , Coronary Vessels/physiopathology , Disease Models, Animal , Endothelium, Vascular/physiopathology , Genome-Wide Association Study , Humans , Ischemia/genetics , Ischemia/metabolism , Ischemia/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , NF-kappa B/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Plaque, Atherosclerotic , Proto-Oncogene Proteins c-akt , Stress, Mechanical
8.
Methods Mol Biol ; 2019: 73-83, 2019.
Article in English | MEDLINE | ID: mdl-31359389

ABSTRACT

Retinoids are reported to regulate vascular growth and remodeling during embryonic development and wound healing. A better understanding of angiogenic mechanisms of retinoids has clinical implication in pathological conditions such as chronic nonhealing wounds. Here, we describe in vitro angiogenesis assays that can be a useful tool to study the role of retinoids and retinoic acid signaling in the regulation of different features of angiogenesis such as tubulogenesis and branching.


Subject(s)
Endothelial Cells/cytology , Fibroblasts/cytology , Neovascularization, Physiologic/drug effects , Retinoids/pharmacology , Animals , Cells, Cultured , Coculture Techniques , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Models, Biological , Receptors, Retinoic Acid/metabolism , Signal Transduction
9.
Nanomedicine ; 18: 391-401, 2019 06.
Article in English | MEDLINE | ID: mdl-30448526

ABSTRACT

Herein, we maximize the labeling efficiency of cardiac progenitor cells (CPCs) using perfluorocarbon nanoparticles (PFCE-NP) and 19F MRI detectability, determine the temporal dynamics of single-cell label uptake, quantify the temporal viability/fluorescence persistence of labeled CPCs in vitro, and implement in vivo, murine cardiac CPC MRI/tracking that could be translatable to humans. FuGENEHD-mediated CPC PFCE-NP uptake is confirmed with flow cytometry/confocal microscopy. Epifluorescence imaging assessed temporal viability/fluorescence (up to 7 days [D]). Nonlocalized murine 19F MRS and cardiac MRI studied label localization in terminal/longitudinal tracking studies at 9.4 T (D1-D8). A 4-8 fold 19F concentration increase is evidenced in CPCs for FuGENE vs. directly labeled cells. Cardiac 19F signals post-CPC injections diminished in vivo to ~31% of their values on D1 by D7/D8. Histology confirmed CPC retention, dispersion, and macrophage-induced infiltration. Intra-cardiac injections of PFCE-NP-labeled CPCs with FuGENE can be visualized/tracked in vivo for the first time with 19F MRI.


Subject(s)
Cell Tracking , Endocytosis , Fluorine/chemistry , Fluorocarbons/metabolism , Magnetic Resonance Imaging , Myocardium/cytology , Nanoparticles/chemistry , Stem Cells/metabolism , Animals , Cell Survival , Female , Fluorescence , Mice, Inbred C57BL , Signal-To-Noise Ratio , Time Factors
10.
Mol Ther Nucleic Acids ; 13: 29-43, 2018 Dec 07.
Article in English | MEDLINE | ID: mdl-30227275

ABSTRACT

Endothelial cell (EC) proliferation is a crucial event in physiological and pathological angiogenesis. MicroRNAs (miRNAs) have emerged as important modulators of the angiogenic switch. Here we conducted high-content screening of a human miRNA mimic library to identify novel regulators of EC growth systematically. Several miRNAs were nominated that enhanced or inhibited EC growth. Of these, we focused on miR-26b, which is a conserved candidate and expressed in multiple human EC types. miR-26b overexpression enhanced EC proliferation, migration, and tube formation, while inhibition of miR-26b suppressed the proliferative and angiogenic capacity of ECs. A combinatory functional small interfering RNA (siRNA) screening of 48 predicted gene targets revealed that miR-26b enhanced EC growth and survival through inhibiting PTEN expression. Local administration of miR-26b mimics promoted the growth of new microvessels in the Matrigel plug model. In the mouse model of hindlimb ischemia, miR-26b was found to be downregulated in endothelium in the first week following ischemia, and local overexpression of miR-26b improved the survival of capillaries and muscle fibers in ischemic muscles. Our findings suggest that miR-26b enhances EC proliferation, survival, and angiogenesis. miR-26b is a potential target for developing novel pro-angiogenic therapeutics in ischemic disease.

11.
Mol Ther ; 26(8): 1996-2007, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29843955

ABSTRACT

Transforming growth factor beta (TGF-ß) is crucial for regulation of the endothelial cell (EC) homeostasis. Perturbation of TGF-ß signaling leads to pathological conditions in the vasculature, causing cardiovascular disease and fibrotic disorders. The TGF-ß pathway is critical in endothelial-to-mesenchymal transition (EndMT), but a gap remains in our understanding of the regulation of TGF-ß and related signaling in the endothelium. This study applied a gain- and loss-of function approach and an in vivo model of skin wound healing to demonstrate that miR-148b regulates TGF-ß signaling and has a key role in EndMT, targeting TGFB2 and SMAD2. Overexpression of miR-148b increased EC migration, proliferation, and angiogenesis, whereas its inhibition promoted EndMT. Cytokine challenge decreased miR-148b levels in ECs while promoting EndMT through the regulation of SMAD2. Finally, in a mouse model of skin wound healing, delivery of miR-148b mimics promoted wound vascularization and accelerated closure. In contrast, inhibition of miR-148b enhanced EndMT in wounds, resulting in impaired wound closure that was reversed by SMAD2 silencing. Together, these results demonstrate for the first time that miR-148b is a key factor controlling EndMT and vascularization. This opens new avenues for therapeutic application of miR-148b in vascular and tissue repair.


Subject(s)
MicroRNAs/genetics , Neovascularization, Physiologic , Signal Transduction , Skin/injuries , Wound Healing , Animals , Cell Movement , Disease Models, Animal , Epithelial-Mesenchymal Transition , Female , Human Umbilical Vein Endothelial Cells , Humans , Mice , Skin/metabolism , Smad2 Protein/metabolism , Transforming Growth Factor beta , Transforming Growth Factor beta2/metabolism
12.
Cardiovasc Res ; 114(10): 1385-1399, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29596571

ABSTRACT

Aims: GTP cyclohydrolase I catalyses the first and rate-limiting reaction in the synthesis of tetrahydrobiopterin (BH4), an essential cofactor for nitric oxide synthases (NOS). Both eNOS and iNOS have been implicated in the progression of atherosclerosis, with opposing effects in eNOS and iNOS knockout mice. However, the pathophysiologic requirement for BH4 in regulating both eNOS and iNOS function, and the effects of loss of BH4 on the progression of atherosclerosis remains unknown. Methods and results: Hyperlipidemic mice deficient in Gch1 in endothelial cells and leucocytes were generated by crossing Gch1fl/flTie2cre mice with ApoE-/- mice. Deficiency of Gch1 and BH4 in endothelial cells and myeloid cells was associated with mildly increased blood pressure. High fat feeding for 6 weeks in Gch1fl/flTie2CreApoE-/- mice resulted in significantly decreased circulating BH4 levels, increased atherosclerosis burden and increased plaque macrophage content. Gch1fl/flTie2CreApoE-/- mice showed hallmarks of endothelial cell dysfunction, with increased aortic VCAM-1 expression and decreased endothelial cell dependent vasodilation. Furthermore, loss of BH4 from pro-inflammatory macrophages resulted in increased foam cell formation and altered cellular redox signalling, with decreased expression of antioxidant genes and increased reactive oxygen species. Bone marrow chimeras revealed that loss of Gch1 in both endothelial cells and leucocytes is required to accelerate atherosclerosis. Conclusion: Both endothelial cell and macrophage BH4 play important roles in the regulation of NOS function and cellular redox signalling in atherosclerosis.


Subject(s)
Aorta/enzymology , Aortic Diseases/enzymology , Atherosclerosis/enzymology , Biopterins/analogs & derivatives , Endothelial Cells/enzymology , GTP Cyclohydrolase/metabolism , Macrophages/enzymology , Animals , Aorta/pathology , Aorta/physiopathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Aortic Diseases/physiopathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Biopterins/metabolism , Blood Pressure , Disease Models, Animal , Disease Progression , Endothelial Cells/pathology , Female , Foam Cells/enzymology , Foam Cells/pathology , GTP Cyclohydrolase/deficiency , GTP Cyclohydrolase/genetics , Macrophages/pathology , Male , Mice, Knockout, ApoE , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/metabolism , Plaque, Atherosclerotic , Reactive Oxygen Species/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Vasoconstriction , Vasodilation
13.
J Pathol ; 242(2): 178-192, 2017 06.
Article in English | MEDLINE | ID: mdl-28299802

ABSTRACT

The functional role of bone morphogenetic protein (BMP) signalling in colorectal cancer (CRC) is poorly defined, with contradictory results in cancer cell line models reflecting the inherent difficulties of assessing a signalling pathway that is context-dependent and subject to genetic constraints. By assessing the transcriptional response of a diploid human colonic epithelial cell line to BMP ligand stimulation, we generated a prognostic BMP signalling signature, which was applied to multiple CRC datasets to investigate BMP heterogeneity across CRC molecular subtypes. We linked BMP and Notch signalling pathway activity and function in human colonic epithelial cells, and normal and neoplastic tissue. BMP induced Notch through a γ-secretase-independent interaction, regulated by the SMAD proteins. In homeostasis, BMP/Notch co-localization was restricted to cells at the top of the intestinal crypt, with more widespread interaction in some human CRC samples. BMP signalling was downregulated in the majority of CRCs, but was conserved specifically in mesenchymal-subtype tumours, where it interacts with Notch to induce an epithelial-mesenchymal transition (EMT) phenotype. In intestinal homeostasis, BMP-Notch pathway crosstalk is restricted to differentiating cells through stringent pathway segregation. Conserved BMP activity and loss of signalling stringency in mesenchymal-subtype tumours promotes a synergistic BMP-Notch interaction, and this correlates with poor patient prognosis. BMP signalling heterogeneity across CRC subtypes and cell lines can account for previous experimental contradictions. Crosstalk between the BMP and Notch pathways will render mesenchymal-subtype CRC insensitive to γ-secretase inhibition unless BMP activation is concomitantly addressed. © 2017 The Authors. Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Bone Morphogenetic Proteins/genetics , Colorectal Neoplasms/genetics , Epithelial-Mesenchymal Transition , Receptors, Notch/genetics , Signal Transduction , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Cohort Studies , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/pathology , Epithelial Cells/pathology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Models, Biological , Phenotype , Prognosis , Receptors, Notch/metabolism , Smad Proteins/genetics , Smad Proteins/metabolism
14.
Mol Ther ; 24(10): 1745-1759, 2016 10.
Article in English | MEDLINE | ID: mdl-27480772

ABSTRACT

Therapeutic angiogenesis is a major goal of regenerative medicine, but no clinically approved small molecule exists that enhances new blood vessel formation. Here we show, using a phenotype-driven high-content imaging screen of an annotated chemical library of 1,280 bioactive small molecules, that the retinoid agonist Tazarotene, enhances in vitro angiogenesis, promoting branching morphogenesis, and tubule remodeling. The proangiogenic phenotype is mediated by retinoic acid receptor but not retinoic X receptor activation, and is characterized by secretion of the proangiogenic factors hepatocyte growth factor, vascular endothelial growth factor, plasminogen activator, urokinase and placental growth factor, and reduced secretion of the antiangiogenic factor pentraxin-3 from adjacent fibroblasts. In vivo, Tazarotene enhanced the growth of mature and functional microvessels in Matrigel implants and wound healing models, and increased blood flow. Notably, in ear punch wound healing model, Tazarotene promoted tissue repair characterized by rapid ear punch closure with normal-appearing skin containing new hair follicles, and maturing collagen fibers. Our study suggests that Tazarotene, an FDA-approved small molecule, could be potentially exploited for therapeutic applications in neovascularization and wound healing.


Subject(s)
Angiogenesis Inducing Agents/administration & dosage , Fibroblasts/cytology , Nicotinic Acids/administration & dosage , Receptors, Retinoic Acid/metabolism , Wound Healing/drug effects , Angiogenesis Inducing Agents/pharmacology , Animals , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Fibroblasts/drug effects , Fibroblasts/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Mice , Nicotinic Acids/pharmacology , Signal Transduction
15.
Hypertension ; 68(3): 749-59, 2016 09.
Article in English | MEDLINE | ID: mdl-27456522

ABSTRACT

Offspring of hypertensive pregnancies are more likely to have microvascular rarefaction and increased blood pressure in later life. We tested the hypothesis that maternal angiogenic profile during a hypertensive pregnancy is associated with fetal vasculogenic capacity and abnormal postnatal microvascular remodeling. Infants (n=255) born after either hypertensive or normotensive pregnancies were recruited for quantification of postnatal dermal microvascular structure at birth and 3 months of age. Vasculogenic cell potential was assessed in umbilical vein endothelial cells from 55 offspring based on in vitro microvessel tube formation and proliferation assays. Maternal angiogenic profile (soluble fms-like tyrosine kinase-1, soluble endoglin, vascular endothelial growth factor, and placental growth factor) was measured from postpartum plasma samples to characterize severity of pregnancy disorder. At birth, offspring born after hypertensive pregnancy had similar microvessel density to those born after a normotensive pregnancy, but during the first 3 postnatal months, they had an almost 2-fold greater reduction in total vessel density (-17.7±16.4% versus -9.9±18.7%; P=0.002). This postnatal loss varied according to the vasculogenic capacity of the endothelial cells of the infant at birth (r=0.49; P=0.02). The degree of reduction in both in vitro and postnatal in vivo vascular development was proportional to levels of antiangiogenic factors in the maternal circulation. In conclusion, our data indicate that offspring born to hypertensive pregnancies have reduced vasculogenic capacity at birth that predicts microvessel density loss over the first 3 postnatal months. Degree of postnatal microvessel reduction is proportional to levels of antiangiogenic factors in the maternal circulation at birth.


Subject(s)
Endothelial Cells/metabolism , Hypertension, Pregnancy-Induced/physiopathology , Microvessels/growth & development , Pregnancy Outcome , Vascular Endothelial Growth Factor Receptor-1/metabolism , Adult , Cohort Studies , Female , Fetal Development/physiology , Humans , Infant , Infant, Newborn , Placenta Growth Factor/metabolism , Pre-Eclampsia/physiopathology , Predictive Value of Tests , Pregnancy , Pregnancy Proteins/blood , Premature Birth/etiology , Premature Birth/physiopathology , Retrospective Studies , Risk Assessment , Umbilical Veins/embryology
16.
Nat Biotechnol ; 34(1): 95-103, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26501955

ABSTRACT

Despite the success of protein kinase inhibitors as approved therapeutics, drug discovery has focused on a small subset of kinase targets. Here we provide a thorough characterization of the Published Kinase Inhibitor Set (PKIS), a set of 367 small-molecule ATP-competitive kinase inhibitors that was recently made freely available with the aim of expanding research in this field and as an experiment in open-source target validation. We screen the set in activity assays with 224 recombinant kinases and 24 G protein-coupled receptors and in cellular assays of cancer cell proliferation and angiogenesis. We identify chemical starting points for designing new chemical probes of orphan kinases and illustrate the utility of these leads by developing a selective inhibitor for the previously untargeted kinases LOK and SLK. Our cellular screens reveal compounds that modulate cancer cell growth and angiogenesis in vitro. These reagents and associated data illustrate an efficient way forward to increasing understanding of the historically untargeted kinome.


Subject(s)
Phosphotransferases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Glycosylation
18.
Circ Res ; 107(2): 283-93, 2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20508179

ABSTRACT

RATIONALE: Notch signaling regulates vascular development. However, the implication of the Notch ligand Delta-like 4 (Dll4) in postischemic angiogenesis remains unclear. OBJECTIVE: We investigated the role of Dll4/Notch signaling in reparative angiogenesis using a mouse model of ischemia. METHODS AND RESULTS: We found Dll4 weakly expressed in microvascular endothelial cells of normoperfused muscles. Conversely, Dll4 is upregulated following ischemia and localized at the forefront of sprouting capillaries. We analyzed the effect of inhibiting endogenous Dll4 by intramuscular injection of an adenovirus encoding the soluble form of Dll4 extracellular domain (Ad-sDll4). Dll4 inhibition caused the formation of a disorganized, low-perfused capillary network in ischemic muscles. This structural abnormality was associated to delayed blood flow recovery and muscle hypoxia and degeneration. Analysis of microvasculature at early stages of repair revealed that Dll4 inhibition enhances capillary sprouting in a chaotic fashion and causes excessive leukocyte infiltration of ischemic muscles. Furthermore, Dll4 inhibition potentiated the elevation of the leukocyte chemoattractant CXCL1 (chemokine [C-X-C motif] ligand 1) following ischemia, without altering peripheral blood levels of stromal cell-derived factor-1 and monocyte chemoattractant protein-1. In cultured human monocytes, Dll4 induces the transcription of Notch target gene Hes-1 and inhibits the basal and tumor necrosis factor-alpha-stimulated production of interleukin-8, the human functional homolog of murine CXCL1. The inhibitory effect of Dll4 on interleukin-8 was abolished by DAPT, a Notch inhibitor, or by coculturing activated human monocytes with Ad-sDll4-infected endothelial cells. CONCLUSIONS: Dll4/Notch interaction is essential for proper reparative angiogenesis. Moreover, Dll4/Notch signaling regulates sprouting angiogenesis and coordinates the interaction between inflammation and angiogenesis under ischemic conditions.


Subject(s)
Endothelial Cells/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Ischemia/metabolism , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Receptors, Notch/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing , Animals , Calcium-Binding Proteins , Cells, Cultured , Chemokine CXCL1/metabolism , Chemotaxis, Leukocyte , Coculture Techniques , Disease Models, Animal , Hindlimb , Humans , Intercellular Signaling Peptides and Proteins/genetics , Interleukin-8/metabolism , Ischemia/diagnostic imaging , Ischemia/genetics , Ischemia/physiopathology , Laser-Doppler Flowmetry , Leukocytes/metabolism , Male , Mice , Neovascularization, Physiologic/genetics , Regeneration , Regional Blood Flow , Signal Transduction/genetics , Time Factors , Transfection , Ultrasonography
19.
Circulation ; 121(15): 1735-45, 2010 Apr 20.
Article in English | MEDLINE | ID: mdl-20368523

ABSTRACT

BACKGROUND: Clinical trials in ischemic patients showed the safety and benefit of autologous bone marrow progenitor cell transplantation. Non-bone marrow progenitor cells with proangiogenic capacities have been described, yet they remain clinically unexploited owing to their scarcity, difficulty of access, and low ex vivo expansibility. We investigated the presence, antigenic profile, expansion capacity, and proangiogenic potential of progenitor cells from the saphenous vein of patients undergoing coronary artery bypass surgery. METHODS AND RESULTS: CD34-positive cells, negative for the endothelial marker von Willebrand factor, were localized around adventitial vasa vasorum. After dissection of the vein from surrounding tissues and enzymatic digestion, CD34-positive/CD31-negative cells were isolated by selective culture, immunomagnetic beads, or fluorescence-assisted cell sorting. In the presence of serum, CD34-positive/CD31-negative cells gave rise to a highly proliferative population that expressed pericyte/mesenchymal antigens together with the stem cell marker Sox2 and showed clonogenic and multilineage differentiation capacities. We called this population "saphenous vein-derived progenitor cells" (SVPs). In culture, SVPs integrated into networks formed by endothelial cells and supported angiogenesis through paracrine mechanisms. Reciprocally, endothelial cell-released factors facilitated SVP migration. These interactive responses were inhibited by Tie-2 or platelet-derived growth factor-BB blockade. Intramuscular injection of SVPs in ischemic limbs of immunodeficient mice improved neovascularization and blood flow recovery. At 14 days after transplantation, proliferating SVPs were still detectable in the recipient muscles, where they established N-cadherin-mediated physical contact with the capillary endothelium. CONCLUSIONS: SVPs generated from human vein CD34-positive/CD31-negative progenitor cells might represent a new therapeutic tool for angiogenic therapy in ischemic patients.


Subject(s)
Adult Stem Cells/cytology , Hematopoietic Stem Cell Transplantation/methods , Ischemia/therapy , Neovascularization, Physiologic/physiology , Saphenous Vein/cytology , Adult , Adult Stem Cells/metabolism , Animals , Antigens, CD34/metabolism , Cell Separation/methods , Cells, Cultured , Clone Cells/cytology , Coronary Artery Bypass , Disease Models, Animal , Flow Cytometry , Graft Survival , Hindlimb/blood supply , Humans , Injections, Intramuscular , Ischemia/pathology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Mutant Strains , Mice, Nude , Platelet Endothelial Cell Adhesion Molecule-1/metabolism
20.
Biochem Soc Trans ; 37(Pt 6): 1221-7, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19909251

ABSTRACT

Notch signalling represents a key pathway essential for normal vascular development. Recently, great attention has been focused on the implication of Notch pathway components in postnatal angiogenesis and regenerative medicine. This paper critically reviews the most recent findings supporting the role of Notch in ischaemia-induced neovascularization. Notch signalling reportedly regulates several steps of the reparative process occurring in ischaemic tissues, including sprouting angiogenesis, vessel maturation, interaction of vascular cells with recruited leucocytes and skeletal myocyte regeneration. Further characterization of Notch interaction with other signalling pathways might help identify novel targets for therapeutic angiogenesis.


Subject(s)
Ischemia/metabolism , Neovascularization, Physiologic/physiology , Receptors, Notch/metabolism , Signal Transduction/physiology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Hypoxia/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Receptors, Notch/genetics , Regeneration/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...