Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Am J Physiol Renal Physiol ; 322(6): F597-F610, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35379003

ABSTRACT

We have previously reported that increased expression and activation of kidney cell complement components play an important role in the pathogenesis of renal scarring. Here, we used floxed green fluorescent protein (GFP)-C5a receptor 1 (C5aR1) knockin mice (GFP-C5ar1fl/fl) and the model of folic acid (FA)-induced kidney injury to define the cell types and potential mechanisms by which increased C5aR1 activation leads to fibrosis. Using flow cytometry and confocal microscopy, we identified macrophages as the major interstitial cell type showing increased expression of C5aR1 in FA-treated mice. C5ar1fl/fl.Lyz2Cre+/- mice, in which C5aR1 has been specifically deleted in lysozyme M-expressing myeloid cells, experienced reduced fibrosis compared with control C5ar1fl/fl mice. Examination of C5aR1-expressing macrophage transcriptomes by gene set enrichment analysis demonstrated that these cells were enriched in pathways corresponding to the complement cascade, collagen formation, and the NABA matrisome, strongly pointing to their critical roles in tissue repair/scarring. Since C5aR1 was also detected in a small population of platelet-derived growth factor receptor-ß+ GFP+ cells, we developed C5ar1fl/fl.Foxd1Cre+/- mice, in which C5aR1 is deleted specifically in pericytes, and found reduced FA-induced fibrosis. Primary cell cultures of platelet-derived growth factor receptor-ß+ pericytes isolated from FA-treated C5ar1fl/fl.Foxd1Cre+/- mice showed reduced secretion of several cytokines, including IL-6 and macrophage inflammatory protein-2, compared with pericytes isolated from FA-treated control GFP-C5ar1fl/fl mice. Collectively, these data imply that C5a/C5aR1 axis activation primarily in interstitial cells contributes to the development of renal fibrosis.NEW & NOTEWORTHY This study used novel green fluorescent protein C5a receptor 1 floxed mice and the model of folic acid-mediated kidney fibrosis to demonstrate the pathogenic role of increased expression of this complement receptor on macrophages.


Subject(s)
Folic Acid , Receptor, Anaphylatoxin C5a , Animals , Cicatrix , Fibrosis , Folic Acid/pharmacology , Green Fluorescent Proteins , Kidney/pathology , Mice , Mice, Knockout , Myeloid Cells/pathology , Receptor, Anaphylatoxin C5a/genetics , Receptors, Platelet-Derived Growth Factor
2.
Nature ; 538(7625): 350-355, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27680706

ABSTRACT

Clostridium difficile toxin B (TcdB) is a critical virulence factor that causes diseases associated with C. difficile infection. Here we carried out CRISPR-Cas9-mediated genome-wide screens and identified the members of the Wnt receptor frizzled family (FZDs) as TcdB receptors. TcdB binds to the conserved Wnt-binding site known as the cysteine-rich domain (CRD), with the highest affinity towards FZD1, 2 and 7. TcdB competes with Wnt for binding to FZDs, and its binding blocks Wnt signalling. FZD1/2/7 triple-knockout cells are highly resistant to TcdB, and recombinant FZD2-CRD prevented TcdB binding to the colonic epithelium. Colonic organoids cultured from FZD7-knockout mice, combined with knockdown of FZD1 and 2, showed increased resistance to TcdB. The colonic epithelium in FZD7-knockout mice was less susceptible to TcdB-induced tissue damage in vivo. These findings establish FZDs as physiologically relevant receptors for TcdB in the colonic epithelium.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Colon/metabolism , Epithelium/metabolism , Frizzled Receptors/metabolism , Animals , Antigens/metabolism , Bacterial Proteins/chemistry , Bacterial Toxins/chemistry , Binding Sites , CHO Cells , CRISPR-Cas Systems , Cell Line , Clostridioides difficile/pathogenicity , Cricetulus , Female , Frizzled Receptors/chemistry , Frizzled Receptors/deficiency , Frizzled Receptors/genetics , Gene Knockout Techniques , Humans , Male , Mice , Mice, Knockout , Oligopeptides/chemistry , Oligopeptides/metabolism , Organoids/metabolism , Protein Domains , Proteoglycans/metabolism , Virulence Factors/metabolism , Wnt Proteins/metabolism
3.
J Biol Chem ; 293(7): 2466-2475, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29196603

ABSTRACT

Sarcomas, and the mesenchymal precursor cells from which they arise, express chondroitin sulfate proteoglycan 4 (NG2/CSPG4). However, NG2/CSPG4's function and its capacity to serve as a therapeutic target in this tumor type are unknown. Here, we used cells from human tumors and a genetically engineered autochthonous mouse model of soft-tissue sarcomas (STSs) to determine NG2/CSPG4's role in STS initiation and growth. Inhibiting NG2/CSPG4 expression in established murine and human STSs decreased tumor volume by almost two-thirds and cell proliferation rate by 50%. NG2/CSPG4 antibody immunotherapy in human sarcomas established as xenografts in mice similarly decreased tumor volume, and expression of a lentivirus blocking NG2/CSPG4 expression inhibited tumor cell proliferation and increased the latency of engraftment. Gene profiling showed that Ng2/Cspg4 deletion altered the expression of genes regulating cell proliferation and apoptosis. Surprisingly, Ng2/Cspg4 deletion at the time of tumor initiation resulted in larger tumors. Gene expression profiling indicated substantial down-regulation of insulin-like growth factor binding protein (Igfbp) genes when Ng2/Cspg4 is depleted at tumor initiation, but not when Ng2/Cspg4 is depleted after tumor initiation. Such differences may have clinical significance, as therapeutic targeting of a signaling pathway such as NG2/CSPG4 may have different effects on cell behavior with tumor progression. NG2/CSPG4 depletion has divergent effects, depending on the developmental stage of sarcoma. In established tumors, IGF signaling is active, and NG2 inhibition targets cell proliferation and apoptosis.


Subject(s)
Antigens/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Membrane Proteins/metabolism , Proteoglycans/metabolism , Sarcoma/metabolism , Sarcoma/physiopathology , Animals , Antigens/genetics , Apoptosis , Cell Line, Tumor , Cell Proliferation , Chondroitin Sulfate Proteoglycans/genetics , Gene Expression Regulation, Neoplastic , Humans , Membrane Proteins/genetics , Mice , Mice, Knockout , Neoplasm Staging , Proteoglycans/genetics , Sarcoma/genetics , Sarcoma/pathology
4.
Adv Exp Med Biol ; 1109: 5-19, 2018.
Article in English | MEDLINE | ID: mdl-30523586

ABSTRACT

Studies of pericytes have been retarded by the lack of appropriate markers for identification of these perivascular mural cells. Use of antibodies against the NG2 proteoglycan as a pericyte marker has greatly facilitated recent studies of pericytes, emphasizing the intimate spatial relationship between pericytes and endothelial cells, allowing more accurate quantification of pericyte/endothelial cell ratios in different vascular beds, and revealing the participation of pericytes throughout all stages of blood vessel formation. The functional importance of NG2 in pericyte biology has been established via NG2 knockdown (in vitro) and knockout (in vivo) strategies that reveal significant deficits in blood vessel formation when NG2 is absent from pericytes. NG2 influences pericyte proliferation and motility by acting as an auxiliary receptor that enhances signaling through integrins and receptor tyrosine kinase growth factor receptors. By acting in a trans orientation, NG2 also activates integrin signaling in closely apposed endothelial cells, leading to enhanced maturation and formation of endothelial cell junctions. NG2 null mice exhibit reduced growth of both mammary and brain tumors that can be traced to deficits in tumor vascularization. Use of Cre-Lox technology to produce pericyte-specific NG2 null mice has revealed specific deficits in tumor vessels that include decreased pericyte ensheathment of endothelial cells, diminished assembly of the vascular basement membrane, reduced vessel patency, and increased vessel leakiness. Interestingly, myeloid-specific NG2 null mice exhibit even larger deficits in tumor vascularization, leading to correspondingly slower tumor growth. Myeloid-specific NG2 null mice are deficient in their ability to recruit macrophages to tumors and other sites of inflammation. This absence of macrophages deprives pericytes of a signal that is crucial for their ability to interact with endothelial cells. The interplay between pericytes, endothelial cells, and macrophages promises to be an extremely fertile area of future study.


Subject(s)
Antigens/chemistry , Pericytes/cytology , Proteoglycans/chemistry , Animals , Endothelial Cells , Inflammation , Macrophages , Mice , Neoplasms , Neovascularization, Pathologic
5.
Am J Physiol Renal Physiol ; 312(3): F516-F532, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28052876

ABSTRACT

We have examined the pathogenic role of increased complement expression and activation during kidney fibrosis. Here, we show that PDGFRß-positive pericytes isolated from mice subjected to obstructive or folic acid injury secrete C1q. This was associated with increased production of proinflammatory cytokines, extracellular matrix components, collagens, and increased Wnt3a-mediated activation of Wnt/ß-catenin signaling, which are hallmarks of myofibroblast activation. Real-time PCR, immunoblots, immunohistochemistry, and flow cytometry analysis performed in whole kidney tissue confirmed increased expression of C1q, C1r, and C1s as well as complement activation, which is measured as increased synthesis of C3 fragments predominantly in the interstitial compartment. Flow studies localized increased C1q expression to PDGFRß-positive pericytes as well as to CD45-positive cells. Although deletion of C1qA did not prevent kidney fibrosis, global deletion of C3 reduced macrophage infiltration, reduced synthesis of C3 fragments, and reduced fibrosis. Clodronate mediated depletion of CD11bF4/80 high macrophages in UUO mice also reduced complement gene expression and reduced fibrosis. Our studies demonstrate local synthesis of complement by both PDGFRß-positive pericytes and CD45-positive cells in kidney fibrosis. Inhibition of complement activation represents a novel therapeutic target to ameliorate fibrosis and progression of chronic kidney disease.


Subject(s)
Complement Activation , Complement C1q/metabolism , Complement C3/metabolism , Kidney Tubules/metabolism , Macrophages/metabolism , Pericytes/metabolism , Renal Insufficiency, Chronic/metabolism , Animals , Cell Communication , Complement C1q/deficiency , Complement C1q/genetics , Complement C1q/immunology , Complement C3/deficiency , Complement C3/genetics , Complement C3/immunology , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Extracellular Matrix Proteins/metabolism , Fibrosis , Folic Acid , Genotype , Inflammation Mediators/metabolism , Kidney Tubules/immunology , Kidney Tubules/pathology , Leukocyte Common Antigens/metabolism , Macrophages/immunology , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Pericytes/immunology , Pericytes/pathology , Phenotype , Receptor, Platelet-Derived Growth Factor beta/metabolism , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/immunology , Renal Insufficiency, Chronic/pathology , Time Factors , Ureteral Obstruction/complications , Wnt Signaling Pathway , Wnt3A Protein/metabolism
6.
Arterioscler Thromb Vasc Biol ; 36(1): 49-59, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26543095

ABSTRACT

OBJECTIVES: Obesity and hyperlipidemia are critical risk factors for atherosclerosis. Because ablation of NG2 proteoglycan in mice leads to hyperlipidemia and obesity, we investigated the impact of NG2 ablation on atherosclerosis in apoE null mice. APPROACH AND RESULTS: Immunostaining indicates that NG2 expression in plaque, primarily by synthetic smooth muscle cells, increases during atherogenesis. NG2 ablation unexpectedly results in decreased (30%) plaque development, despite aggravated obesity and hyperlipidemia. Mechanistic studies reveal that NG2-positive plaque synthetic smooth muscle cells in culture can sequester low-density lipoprotein to enhance foam-cell formation, processes in which NG2 itself plays direct roles. In agreement with these observations, low-density lipoprotein retention and lipid accumulation in the NG2/ApoE knockout aorta is 30% less than that seen in the control aorta. CONCLUSIONS: These results indicate that synthetic smooth muscle cell-dependent low-density lipoprotein retention and foam cell formation outweigh obesity and hyperlipidemia in promoting mouse atherogenesis. Our study sheds new light on the role of synthetic smooth muscle cells during atherogenesis. Blocking plaque NG2 or altering synthetic smooth muscle cells function may be promising therapeutic strategies for atherosclerosis.


Subject(s)
Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Foam Cells/metabolism , Lipoproteins, LDL/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Proteoglycans/deficiency , Animals , Antigens/genetics , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cells, Cultured , Diet, High-Fat , Disease Models, Animal , Female , Foam Cells/pathology , Hyperlipidemias/genetics , Hyperlipidemias/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Obesity/genetics , Obesity/metabolism , Plaque, Atherosclerotic , Proteoglycans/genetics
7.
Microcirculation ; 23(2): 122-33, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26465118

ABSTRACT

The NG2 proteoglycan promotes tumor growth as a component of both tumor and stromal cells. Using intracranial, NG2-negative B16F10 melanomas, we have investigated the importance of PC and Mac NG2 in brain tumor progression. Reduced melanoma growth in Mac-NG2ko and PC-NG2ko mice demonstrates the importance of NG2 in both stromal compartments. In each genotype, the loss of PC-endothelial cell interaction diminishes the formation of endothelial junctions and assembly of the basal lamina. Tumor vessels in Mac-NG2ko mice have smaller diameters, reduced patency, and increased leakiness compared to PC-NG2ko mice, thus decreasing tumor blood supply and increasing hypoxia. While the reduced PC interaction with endothelial cells in PC-NG2ko mice results from the loss of PC activation of ß1 integrin signaling in endothelial cells, reduced PC-endothelial cell interaction in Mac-NG2ko mice results from 90% reduced Mac recruitment. The absence of Mac-derived signals in Mac-NG2ko mice causes the loss of PC association with endothelial cells. Reduced Mac recruitment may be due to diminished activation of integrins in the absence of NG2, causing decreased Mac interaction with endothelial adhesion molecules that are needed for extravasation. These results reflect the complex interplay that occurs between Mac, PC, and endothelial cells during tumor vascularization.


Subject(s)
Antigens/metabolism , Brain Neoplasms/metabolism , Macrophages/metabolism , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/metabolism , Pericytes/metabolism , Proteoglycans/metabolism , Animals , Antigens/genetics , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Macrophages/pathology , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Pericytes/pathology , Proteoglycans/genetics
8.
Adv Funct Mater ; 26(2): 267-276, 2016 Jan 13.
Article in English | MEDLINE | ID: mdl-27441036

ABSTRACT

The rapid development of fluorescence imaging technologies requires concurrent improvements in the performance of fluorescent probes. Quantum dots have been extensively used as an imaging probe in various research areas because of their inherent advantages based on unique optical and electronic properties. However, their clinical translation has been limited by the potential toxicity especially from cadmium. Here, a versatile bioimaging probe is developed by using highly luminescent cadmium-free CuInSe2/ZnS core/shell quantum dots conjugated with CGKRK (Cys-Gly-Lys-Arg-Lys) tumor-targeting peptides. This probe exhibits excellent photostability, reasonably long circulation time, minimal toxicity, and strong tumor-specific homing property. The most important feature of this probe is that it shows distinctive versatility in tumor-targeted multimodal imaging including near-infrared, time-gated, and two-photon imaging in different tumor models. In a glioblastoma mouse model, the targeted probe clearly denotes tumor boundaries and positively labels a population of diffusely infiltrating tumor cells, suggesting its utility in precise tumor detection during surgery. This work lays a foundation for potential clinical translation of the probe.

9.
J Neurosci ; 34(49): 16369-84, 2014 Dec 03.
Article in English | MEDLINE | ID: mdl-25471575

ABSTRACT

NG2 is purportedly one of the most growth-inhibitory chondroitin sulfate proteoglycans (CSPGs) produced after spinal cord injury. Nonetheless, once the severed axon tips dieback from the lesion core into the penumbra they closely associate with NG2+ cells. We asked if proteoglycans play a role in this tight cell-cell interaction and whether overadhesion upon these cells might participate in regeneration failure in rodents. Studies using varying ratios of CSPGs and adhesion molecules along with chondroitinase ABC, as well as purified adult cord-derived NG2 glia, demonstrate that CSPGs are involved in entrapping neurons. Once dystrophic axons become stabilized upon NG2+ cells, they form synaptic-like connections both in vitro and in vivo. In NG2 knock-out mice, sensory axons in the dorsal columns dieback further than their control counterparts. When axons are double conditioned to enhance their growth potential, some traverse the lesion core and express reduced amounts of synaptic proteins. Our studies suggest that proteoglycan-mediated entrapment upon NG2+ cells is an additional obstacle to CNS axon regeneration.


Subject(s)
Antigens/physiology , Axons/physiology , Cell Communication/physiology , Nerve Regeneration/physiology , Proteoglycans/physiology , Spinal Cord Injuries/physiopathology , Synapses/physiology , Animals , Antigens/genetics , Axons/ultrastructure , Cell Tracking , Cells, Cultured , Chondroitin Sulfate Proteoglycans/physiology , Fibronectins/physiology , Ganglia, Spinal/physiopathology , Ganglia, Spinal/ultrastructure , Integrin beta1/physiology , Laminin/physiology , Mice , Mice, Knockout , Nerve Degeneration/physiopathology , Proteoglycans/genetics
10.
J Neuroinflammation ; 12: 161, 2015 Sep 04.
Article in English | MEDLINE | ID: mdl-26338007

ABSTRACT

BACKGROUND: The NG2 proteoglycan is expressed by several cell types in demyelinated lesions and has important effects on the biology of these cells. Here we determine the cell-type-specific roles of NG2 in the oligodendrocyte progenitor cell (OPC) and myeloid cell contributions to demyelination and remyelination. METHODS: We have used Cre-Lox technology to dissect the cell-type-specific contributions of NG2 to myelin damage and repair. Demyelination is induced by microinjection of 1 % lysolecithin into the spinal cord white matter of control, OPC-specific NG2-null (OPC-NG2ko), and myeloid-specific NG2-null (My-NG2ko) mice. The status of OPCs, myeloid cells, axons, and myelin is assessed by light, immunofluorescence, confocal, and electron microscopy. RESULTS: In OPC-NG2ko mice 1 week after lysolecithin injection, the OPC mitotic index is reduced by 40 %, resulting in 25 % fewer OPCs at 1 week and a 28 % decrease in mature oligodendrocytes at 6 weeks post-injury. The initial demyelinated lesion size is not affected in OPC-NG2ko mice, but lesion repair is delayed by reduced production of oligodendrocytes. In contrast, both the initial extent of demyelination and the kinetics of lesion repair are decreased in My-NG2ko mice. Surprisingly, the OPC mitotic index at 1 week post-injury is also reduced (by 48 %) in My-NG2ko mice, leading to a 35 % decrease in OPCs at 1 week and a subsequent 34 % reduction in mature oligodendrocytes at 6 weeks post-injury. Clearance of myelin debris is also reduced by 40 % in My-NG2ko mice. Deficits in myelination detected by immunostaining for myelin basic protein are confirmed by toluidine blue staining and by electron microscopy. In addition to reduced myelin repair, fewer axons are found in 6-week lesions in both OPC-NG2ko and My-NG2ko mice, emphasizing the importance of myelination for neuron survival. CONCLUSIONS: Reduced generation of OPCs and oligodendrocytes in OPC-NG2ko mice correlates with reduced myelin repair. Diminished demyelination in My-NG2ko mice may stem from a reduction (approximately 70 %) in myeloid cell recruitment to lesions. Reduced macrophage/microglia numbers may then result in decreased myelin repair via diminished clearance of myelin debris and reduced stimulatory effects on OPCs.


Subject(s)
Antigens/metabolism , Demyelinating Autoimmune Diseases, CNS/pathology , Myeloid Cells/pathology , Oligodendroglia/pathology , Proteoglycans/metabolism , Recovery of Function/physiology , Spinal Cord/pathology , Animals , Antigens/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Marrow Transplantation , Calcium-Binding Proteins/metabolism , Cell Differentiation/physiology , Demyelinating Autoimmune Diseases, CNS/chemically induced , Demyelinating Autoimmune Diseases, CNS/surgery , Disease Models, Animal , Lysophosphatidylcholines/toxicity , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Myelin Sheath/metabolism , Myelin Sheath/pathology , Myelin Sheath/ultrastructure , Myeloid Cells/ultrastructure , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/metabolism , Phagocytosis/physiology , Proteoglycans/genetics , Stem Cells/metabolism , Stem Cells/ultrastructure
11.
Am J Pathol ; 184(2): 541-55, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24326256

ABSTRACT

Blood-retinal barrier (BRB) breakdown and related vascular changes are implicated in several ocular diseases. The molecules and mechanisms regulating BRB integrity and pathophysiology are not fully elucidated. Caveolin-1 (Cav-1) ablation results in loss of caveolae and microvascular pathologies, but the role of Cav-1 in the retina is largely unknown. We examined BRB integrity and vasculature in Cav-1 knockout mice and found a significant increase in BRB permeability, compared with wild-type controls, with branch veins being frequent sites of breakdown. Vascular hyperpermeability occurred without apparent alteration in junctional proteins. Such hyperpermeability was not rescued by inhibiting eNOS activity. Veins of Cav-1 knockout retinas exhibited additional pathological features, including i) eNOS-independent enlargement, ii) altered expression of mural cell markers (eg, down-regulation of NG2 and up-regulation of αSMA), and iii) dramatic alterations in mural cell phenotype near the optic nerve head. We observed a significant NO-dependent increase in retinal artery diameter in Cav-1 knockout mice, suggesting that Cav-1 plays a role in autoregulation of resistance vessels in the retina. These findings implicate Cav-1 in maintaining BRB integrity in retinal vasculature and suggest a previously undefined role in the retinal venous system and associated mural cells. Our results are relevant to clinically significant retinal disorders with vascular pathologies, including diabetic retinopathy, uveoretinitis, and primary open-angle glaucoma.


Subject(s)
Blood-Retinal Barrier/metabolism , Blood-Retinal Barrier/pathology , Caveolin 1/deficiency , Retinal Vein/metabolism , Retinal Vein/pathology , Animals , Biomarkers/metabolism , Blood-Retinal Barrier/enzymology , Blood-Retinal Barrier/ultrastructure , Caveolin 1/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type III/metabolism , Permeability , Phenotype , Protein Transport , Retinal Vein/enzymology , Retinal Vein/ultrastructure , Tight Junction Proteins/metabolism
12.
Angiogenesis ; 17(1): 61-76, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23925489

ABSTRACT

The NG2 proteoglycan stimulates the proliferation and migration of various immature cell types, including pericytes. However, the role of NG2 in mediating pericyte/endothelial cell interaction has been less clear. In this study, we show that pericyte-specific NG2 ablation causes several structural deficits in blood vessels in intracranial B16F10 melanomas, including decreased pericyte ensheathment of endothelial cells, diminished formation of endothelial junctions, and reduced assembly of the vascular basal lamina. These deficits result in decreased tumor vessel patency, increased vessel leakiness, and increased intratumoral hypoxia. NG2-dependent mechanisms of pericyte interaction with endothelial cells are further explored in pericyte/endothelial cell co-cultures. siRNA-mediated NG2 knockdown in pericytes leads to reduced formation of pericyte/endothelial networks, reduced formation of ZO-1 positive endothelial cell junctions, and increased permeability of endothelial cell monolayers. We also show that NG2 knockdown results in loss of ß1 integrin activation in endothelial cells, revealing a mechanism for NG2-dependent cross talk between pericytes and endothelial cells.


Subject(s)
Antigens/metabolism , Integrin beta1/metabolism , Melanoma/blood supply , Melanoma/metabolism , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/metabolism , Pericytes/metabolism , Proteoglycans/metabolism , Animals , Antigens/genetics , Cell Line, Tumor , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Integrin beta1/genetics , Integrins , Intercellular Junctions/genetics , Intercellular Junctions/metabolism , Intercellular Junctions/pathology , Melanoma/genetics , Melanoma/mortality , Mice , Neoplasm Proteins/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Pericytes/pathology , Proteoglycans/genetics , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism
13.
Nature ; 454(7200): 104-8, 2008 Jul 03.
Article in English | MEDLINE | ID: mdl-18480752

ABSTRACT

Understanding the origins and roles of cardiac progenitor cells is important for elucidating the pathogenesis of congenital and acquired heart diseases. Moreover, manipulation of cardiac myocyte progenitors has potential for cell-based repair strategies for various myocardial disorders. Here we report the identification in mouse of a previously unknown cardiac myocyte lineage that derives from the proepicardial organ. These progenitor cells, which express the T-box transcription factor Tbx18, migrate onto the outer cardiac surface to form the epicardium, and then make a substantial contribution to myocytes in the ventricular septum and the atrial and ventricular walls. Tbx18-expressing cardiac progenitors also give rise to cardiac fibroblasts and coronary smooth muscle cells. The pluripotency of Tbx18 proepicardial cells provides a theoretical framework for applying these progenitors to effect cardiac repair and regeneration.


Subject(s)
Cell Lineage , Myocardium/cytology , Myocytes, Cardiac/cytology , Pericardium/cytology , Pericardium/metabolism , Stem Cells/cytology , T-Box Domain Proteins/metabolism , Animals , Cell Differentiation , Gene Expression Regulation, Developmental , Heart/growth & development , Lac Operon/genetics , Mice , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Smooth Muscle/metabolism , Stem Cells/metabolism , T-Box Domain Proteins/genetics
14.
Angiogenesis ; 16(2): 309-27, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23124902

ABSTRACT

Sprouting of angiogenic perivascular cells is thought to be highly dependent upon autocrine and paracrine growth factor stimulation. Accordingly, we report that corneal angiogenesis induced by ectopic FGF implantation is strongly impaired in NG2/CSPG4 proteoglycan (PG) null mice known to harbour a putative deficit in pericyte proliferation/mobilization. Conversely, no significant differences were seen between wild type and knockout corneas when VEGF was used as an angiocrine factor. Perturbed responsiveness of NG2-deficient pericytes to paracrine and autocrine stimulation by several FGFs could be confirmed in cells isolated from NG2 null mice, while proliferation induced by other growth factors was equivalent in wild type and knockout cells. Identical results were obtained after siRNA-mediated knock-down of NG2 in human smooth muscle-like cell lines, as also demonstrated by the decreased levels of FGF receptor phosphorylation detected in these NG2 deprived cells. Binding assays with recombinant proteins and molecular interactions examined on live cells asserted that FGF-2 bound to NG2 in a glycosaminoglycan-independent, core protein-mediated manner and that the PG was alone capable of retaining FGF-2 on the cell membrane for subsequent receptor presentation. The use of dominant-negative mutant cells, engineered by combined transduction of NG2 deletion constructs and siRNA knock-down of the endogenous PG, allowed us to establish that the FGF co-receptor activity of NG2 is entirely mediated by its extracellular portion. In fact, forced overexpression of the NG2 ectodomain in human smooth muscle-like cells increased their FGF-2-induced mitosis and compensated for low levels of FGF receptor surface expression, in a manner equivalent to that produced by overexpression of the full-length NG2. Upon FGF binding, the cytoplasmic domain of NG2 is phosphorylated, but there is no evidence that this event elicits signal transductions that could bypass the FGFR-mediated ones. Pull-down experiments, protein-protein binding assays and flow cytometry FRET coherently revealed an elective ligand-independent association of NG2 with FGFR1 and FGFR3. The NG2 cooperation with these receptors was also corroborated functionally by the outcome of FGF-2 treatments of cells engineered to express diverse NG2/FGFR combinations. Comprehensively, the findings suggest that perivascular NG2 may serve as a dual modulator of the availability/accessibility of FGF at the cell membrane, as well as the resulting FGFR transducing activity.


Subject(s)
Antigens/metabolism , Fibroblast Growth Factors/metabolism , Mitogens/metabolism , Pericytes/metabolism , Proteoglycans/metabolism , Animals , Cornea/blood supply , Fluorescence Resonance Energy Transfer , Fluorescent Antibody Technique , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Signal Transduction
15.
Am J Pathol ; 180(3): 1145-1158, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22200614

ABSTRACT

To investigate the importance of the vascular basal lamina in tumor blood vessel morphogenesis and function, we compared vessel development, vessel function, and progression of B16F10 melanoma tumors in the brains of wild-type and collagen VI-null mice. In 7-day tumors in the absence of collagen VI, the width of the vascular basal lamina was reduced twofold. Although the ablation of collagen VI did not alter the abundance of blood vessels, a detailed analysis of the number of either pericytes or endothelial cells (or pericyte coverage of endothelial cells) showed that collagen VI-dependent defects during the assembly of the basal lamina have negative effects on both pericyte maturation and the sprouting and survival of endothelial cells. As a result of these deficits, vessel patency was reduced by 25%, and vessel leakiness was increased threefold, resulting in a 10-fold increase in tumor hypoxia along with a fourfold increase in hypoxia-inducible factor-1α expression. In 12-day collagen VI-null tumors, vascular endothelial growth factor expression was increased throughout the tumor stroma, in contrast to the predominantly vascular pattern of vascular endothelial growth factor expression in wild-type tumors. Vessel size was correspondingly reduced in 12-day collagen VI-null tumors. Overall, these vascular deficits produced a twofold decrease in tumor volume in collagen VI-null mice, confirming that collagen VI-dependent basal lamina assembly is a critical aspect of vessel development.


Subject(s)
Brain Neoplasms/blood supply , Collagen Type VI/physiology , Melanoma/blood supply , Animals , Apoptosis , Basement Membrane/pathology , Brain Neoplasms/pathology , Cell Hypoxia/physiology , Collagen Type VI/deficiency , Disease Progression , Endothelial Cells , Endothelium, Vascular/pathology , Hypoxia-Inducible Factor 1/metabolism , Melanoma/pathology , Mice , Mice, Inbred C57BL , Necrosis , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Vascular Endothelial Growth Factor A/metabolism , Vascular Patency
16.
J Vasc Res ; 50(2): 134-44, 2013.
Article in English | MEDLINE | ID: mdl-23258211

ABSTRACT

Restenosis is a major complication of coronary angioplasty, at least partly due to the fact that the origin and identity of contributing cell types are not well understood. In this study, we have investigated whether pericyte-like cells or mesenchymal stem cells (MSCs) from the adventitia contribute to restenosis. We demonstrate that while cells expressing the pericyte markers NG2, platelet-derived growth factor receptor ß, and CD146 are rare in the adventitia of uninjured mouse femoral arteries, following injury their numbers strongly increase. Some of these adventitial pericyte-like cells acquire a more MSC-like phenotype (CD90+ and CD29+ are up-regulated) and also appear in the restenotic neointima. Via bone marrow transplantation and ex vivo artery culture approaches, we demonstrate that the pericyte-like MSCs of the injured femoral artery are not derived from the bone marrow, but originate in the adventitia itself mainly via the proliferation of resident pericyte-like cells. In summary, we have identified a population of resident adventitial pericyte-like cells or MSCs that contribute to restenosis following arterial injury. These cells are different from myofibroblasts, smooth muscle cells, and other progenitor populations that have been shown to participate in the restenotic process.


Subject(s)
Adventitia/pathology , Arterial Occlusive Diseases/physiopathology , Femoral Artery/injuries , Mesenchymal Stem Cells/physiology , Neointima/physiopathology , Pericytes/physiology , Animals , Antigens/analysis , Antigens/biosynthesis , Antigens/genetics , Antigens, CD/analysis , Aorta, Thoracic/cytology , Bone Marrow Transplantation , Cell Lineage , Constriction, Pathologic , Femoral Artery/pathology , Gene Expression Profiling , Genes, Reporter , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Proteoglycans/analysis , Proteoglycans/biosynthesis , Proteoglycans/genetics , Radiation Chimera , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Receptor, Platelet-Derived Growth Factor beta/genetics , Recurrence
17.
Breast Cancer Res ; 14(2): R67, 2012 Apr 24.
Article in English | MEDLINE | ID: mdl-22531600

ABSTRACT

INTRODUCTION: The neuron-glial antigen 2 (NG2) proteoglycan promotes pericyte recruitment and mediates pericyte interaction with endothelial cells. In the absence of NG2, blood vessel development is negatively impacted in several pathological models. Our goal in this study was to determine the effect of NG2 ablation on the early development and function of blood vessels in mammary tumors in the mammary tumor virus-driven polyoma middle T (MMTV-PyMT) transgenic mouse, and to correlate these vascular changes with alterations in mammary tumor growth. METHODS: Three different tumor paradigms (spontaneous tumors, transplanted tumors, and orthotopic allografts of tumor cell lines) were used to investigate the effects of NG2 ablation on breast cancer progression in the MMTV-PyMT transgenic mouse. In addition to examining effects of NG2 ablation on mammary tumor growth, we also investigated effects on the structure and function of tumor vasculature. RESULTS: Ablation of NG2 led to reduced early progression of spontaneous, transplanted, and orthotopic allograft mammary tumors. NG2 was not expressed by the mammary tumor cells themselves, but instead was found on three components of the tumor stroma. Microvascular pericytes, myeloid cells, and adipocytes were NG2-positive in both mouse and human mammary tumor stroma. The effect of NG2 on tumor progression therefore must be stromal in nature. Ablation of NG2 had several negative effects on early development of the mammary tumor vasculature. In the absence of NG2, pericyte ensheathment of endothelial cells was reduced, along with reduced pericyte maturation, reduced sprouting of endothelial cells, reduced assembly of the vascular basal lamina, and reduced tumor vessel diameter. These early deficits in vessel structure are accompanied by increased vessel leakiness, increased tumor hypoxia, and decreased tumor growth. NG2 ablation also diminishes the number of tumor-associated and TEK tyrosine kinase endothelial (Tie2) expressing macrophages in mammary tumors, providing another possible mechanism for reducing tumor vascularization and growth. CONCLUSIONS: These results emphasize the importance of NG2 in mediating pericyte/endothelial cell communication that is required for proper vessel maturation and function. In the absence of normal pericyte/endothelial cell interaction, poor vascular function results in diminished early progression of mammary tumors.


Subject(s)
Antigens/genetics , Blood Vessels/pathology , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/pathology , Neovascularization, Pathologic/genetics , Proteoglycans/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adipocytes/metabolism , Adipocytes/pathology , Animals , Antigens/metabolism , Antigens, Polyomavirus Transforming/genetics , Blood Vessels/metabolism , Female , Humans , Macrophages/metabolism , Macrophages/pathology , Male , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/physiology , Mammary Neoplasms, Experimental/genetics , Mammary Tumor Virus, Mouse/genetics , Mammary Tumor Virus, Mouse/pathogenicity , Mice , Mice, Transgenic , Proteoglycans/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, TIE-2 , Stromal Cells/metabolism , Stromal Cells/pathology , Xenograft Model Antitumor Assays
18.
J Cell Biol ; 178(1): 155-65, 2007 Jul 02.
Article in English | MEDLINE | ID: mdl-17591920

ABSTRACT

Two distinct Thr phosphorylation events within the cytoplasmic domain of the NG2 proteoglycan help regulate the cellular balance between proliferation and motility. Protein kinase Calpha mediates the phosphorylation of NG2 at Thr2256, resulting in enhanced cell motility. Extracellular signal-regulated kinase phosphorylates NG2 at Thr2314, stimulating cell proliferation. The effects of NG2 phosphorylation on proliferation and motility are dependent on beta1-integrin activation. Differential cell surface localization of the two distinctly phosphorylated forms of NG2 may be the mechanism by which the NG2-beta1-integrin interaction promotes proliferation in one case and motility in the other. NG2 phosphorylated at Thr2314 colocalizes with beta1-integrin on microprotrusions from the apical cell surface. In contrast, NG2 phosphorylated at Thr2256 colocalizes with beta1-integrin on lamellipodia at the leading edges of cells. Thus, phosphorylation and the resulting site of NG2-integrin localization may determine the specific downstream effects of integrin signaling.


Subject(s)
Antigens/metabolism , Cell Movement , Cell Proliferation , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase C-alpha/metabolism , Proteoglycans/metabolism , Adenoviridae/genetics , Amino Acid Sequence , Animals , Antigens/chemistry , Antigens/genetics , Astrocytoma/pathology , Cell Line, Tumor , Conserved Sequence , DNA, Complementary/genetics , Genetic Vectors , Humans , Hybridomas/cytology , Integrin beta1/metabolism , Melanoma/pathology , Molecular Sequence Data , Mutation , Phosphorylation , Protein Structure, Tertiary , Proteoglycans/chemistry , Proteoglycans/genetics , Pseudopodia/metabolism , Rats , Recombinant Fusion Proteins/metabolism , Threonine/metabolism
19.
BMC Physiol ; 12: 7, 2012 Jun 21.
Article in English | MEDLINE | ID: mdl-22720777

ABSTRACT

BACKGROUND: Observations in our laboratory provide evidence of vascular islands, defined as disconnected endothelial cell segments, in the adult microcirculation. The objective of this study was to determine if vascular islands are involved in angiogenesis during microvascular network growth. RESULTS: Mesenteric tissues, which allow visualization of entire microvascular networks at a single cell level, were harvested from unstimulated adult male Wistar rats and Wistar rats 3 and 10 days post angiogenesis stimulation by mast cell degranulation with compound 48/80. Tissues were immunolabeled for PECAM and BRDU. Identification of vessel lumens via injection of FITC-dextran confirmed that endothelial cell segments were disconnected from nearby patent networks. Stimulated networks displayed increases in vascular area, length density, and capillary sprouting. On day 3, the percentage of islands with at least one BRDU-positive cell increased compared to the unstimulated level and was equal to the percentage of capillary sprouts with at least one BRDU-positive cell. At day 10, the number of vascular islands per vascular area dramatically decreased compared to unstimulated and day 3 levels. CONCLUSIONS: These results show that vascular islands have the ability to proliferate and suggest that they are able to incorporate into the microcirculation during the initial stages of microvascular network growth.


Subject(s)
Capillaries/cytology , Endothelial Cells/cytology , Microcirculation/physiology , Microvessels/cytology , Neovascularization, Physiologic/physiology , Animals , Capillaries/growth & development , Cell Growth Processes/physiology , Male , Mast Cells/physiology , Mesentery/blood supply , Neovascularization, Pathologic/physiopathology , Rats , Rats, Wistar
20.
Proc Natl Acad Sci U S A ; 106(42): 17992-7, 2009 Oct 20.
Article in English | MEDLINE | ID: mdl-19815532

ABSTRACT

The formation of oligodendrocytes (oligodendrogenesis) and myelin is regulated by several neurotrophic factors. Strategies to increase the level of these trophic molecules may facilitate repair in demyelinating conditions, such as multiple sclerosis (MS). Because leukocytes are a source of neurotrophic factors, and as glatiramer acetate (GA) generates T helper 2 (Th2) lymphocytes that are not known to be harmful, we tested the hypothesis that GA regulates oligodendrogenesis and myelin formation. First, we generated GA-reactive Th2 cells and determined that they produced transcripts for neurotrophic factors, including insulin-like growth factor-1 (IGF-1). The conditioned medium from GA-reactive T cells elevated IGF-1 protein and promoted the formation of oligodendrocyte precursor cells (OPCs) from embryonic brain-derived forebrain cells in culture. We next subjected mice to lysolecithin-induced demyelination of the spinal cord. At 7 days after the insult, the number of OPCs in the demyelinated dorsal column was higher than that in uninjured controls, and was further increased by the daily s.c. injection with GA. Increased OPC generation by GA was associated temporally with the elevation of IGF-1 and brain-derived neurotrophic factor (BDNF) in the spinal cord. Finally, the resultant remyelination at 28 days was higher in mice treated with GA during the first 7 days of injury compared with vehicle controls. These results indicate that GA promotes oligodendrogenesis and remyelination through mechanisms that involve the elevation of growth factors conducive for repair.


Subject(s)
Multiple Sclerosis/drug therapy , Myelin Sheath/drug effects , Neurogenesis/drug effects , Oligodendroglia/drug effects , Peptides/pharmacology , Animals , Culture Media, Conditioned , Cytokines/metabolism , Glatiramer Acetate , Humans , Immunosuppressive Agents/pharmacology , In Vitro Techniques , Inflammation Mediators/metabolism , Lysophosphatidylcholines/toxicity , Male , Mice , Mice, Inbred C57BL , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , Myelin Sheath/immunology , Myelin Sheath/physiology , Nerve Growth Factors/biosynthesis , Nerve Growth Factors/genetics , Neurogenesis/physiology , Oligodendroglia/cytology , Oligodendroglia/immunology , Oligodendroglia/physiology , Spinal Cord/drug effects , Spinal Cord/pathology , Spinal Cord/physiopathology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/physiology
SELECTION OF CITATIONS
SEARCH DETAIL