Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 24
1.
JCI Insight ; 8(22)2023 Nov 22.
Article En | MEDLINE | ID: mdl-37991018

We previously established that vascular smooth muscle-derived adventitial progenitor cells (AdvSca1-SM) preferentially differentiate into myofibroblasts and contribute to fibrosis in response to acute vascular injury. However, the role of these progenitor cells in chronic atherosclerosis has not been defined. Using an AdvSca1-SM cell lineage tracing model, scRNA-Seq, flow cytometry, and histological approaches, we confirmed that AdvSca1-SM-derived cells localized throughout the vessel wall and atherosclerotic plaques, where they primarily differentiated into fibroblasts, smooth muscle cells (SMC), or remained in a stem-like state. Krüppel-like factor 4 (Klf4) knockout specifically in AdvSca1-SM cells induced transition to a more collagen-enriched fibroblast phenotype compared with WT mice. Additionally, Klf4 deletion drastically modified the phenotypes of non-AdvSca1-SM-derived cells, resulting in more contractile SMC and atheroprotective macrophages. Functionally, overall plaque burden was not altered with Klf4 deletion, but multiple indices of plaque composition complexity, including necrotic core area, macrophage accumulation, and fibrous cap thickness, were reduced. Collectively, these data support that modulation of AdvSca1-SM cells through KLF4 depletion confers increased protection from the development of potentially unstable atherosclerotic plaques.


Plaque, Atherosclerotic , Mice , Animals , Plaque, Atherosclerotic/pathology , Kruppel-Like Factor 4 , Myocytes, Smooth Muscle/pathology , Stem Cells/pathology , Muscle, Smooth/pathology
2.
Arterioscler Thromb Vasc Biol ; 43(11): 2223-2230, 2023 11.
Article En | MEDLINE | ID: mdl-37706321

BACKGROUND: In recent years, fate-mapping lineage studies in mouse models have led to major advances in vascular biology by allowing investigators to track specific cell populations in vivo. One of the most frequently used lineage tracing approaches involves tamoxifen-inducible CreERT-LoxP systems. However, tamoxifen treatment can also promote effects independent of Cre recombinase activation, many of which have not been fully explored. METHODS: To elucidate off-target effects of tamoxifen, male and female mice were either unmanipulated or injected with tamoxifen or corn oil. All mice received PCSK9 (proprotein convertase subtilisin/kexin type 9)-AAV (adeno-associated virus) injections and a modified Western diet to induce hypercholesterolemia. After 2 weeks, serum cholesterol and liver morphology were assessed. To determine the duration of any tamoxifen effects in long-term atherosclerosis experiments, mice received either 12 days of tamoxifen at baseline or 12 days plus 2 sets of 5-day tamoxifen boosters; all mice received PCSK9-AAV injections and a modified Western diet to induce hypercholesterolemia. After 24 weeks, serum cholesterol and aortic sinus plaque burden were measured. RESULTS: After 2 weeks of atherogenic treatment, mice injected with tamoxifen demonstrated significantly reduced serum cholesterol levels compared with uninjected- or corn oil-treated mice. However, there were no differences in PCSK9-mediated knockdown of LDL (low-density lipoprotein) receptors between the groups. Additionally, tamoxifen-treated mice exhibited significantly increased hepatic lipid accumulation compared with the other groups. Finally, the effects of tamoxifen remained for at least 8 weeks after completion of injections, with mice demonstrating persistent decreased serum cholesterol and impaired atherosclerotic plaque formation. CONCLUSIONS: In this study, we establish that tamoxifen administration results in decreased serum cholesterol, decreased plaque formation, and increased hepatic lipid accumulation. These alterations represent significant confounding variables in atherosclerosis research, and we urge future investigators to take these findings into consideration when planning and executing their own atherosclerosis experiments.


Atherosclerosis , Hypercholesterolemia , Plaque, Atherosclerotic , Male , Female , Mice , Animals , Proprotein Convertase 9/metabolism , Hypercholesterolemia/drug therapy , Corn Oil , Atherosclerosis/chemically induced , Atherosclerosis/genetics , Atherosclerosis/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , Cholesterol , Mice, Inbred C57BL
3.
bioRxiv ; 2023 Jul 19.
Article En | MEDLINE | ID: mdl-37503181

We previously established that vascular smooth muscle-derived adventitial progenitor cells (AdvSca1-SM) preferentially differentiate into myofibroblasts and contribute to fibrosis in response to acute vascular injury. However, the role of these progenitor cells in chronic atherosclerosis has not been defined. Using an AdvSca1-SM lineage tracing model, scRNA-Seq, flow cytometry, and histological approaches, we confirmed that AdvSca1-SM cells localize throughout the vessel wall and atherosclerotic plaques, where they primarily differentiate into fibroblasts, SMCs, or remain in a stem-like state. Klf4 knockout specifically in AdvSca1-SM cells induced transition to a more collagen-enriched myofibroblast phenotype compared to WT mice. Additionally, Klf4 depletion drastically modified the phenotypes of non-AdvSca1-SM-derived cells, resulting in more contractile SMCs and atheroprotective macrophages. Functionally, overall plaque burden was not altered with Klf4 depletion, but multiple indices of plaque vulnerability were reduced. Collectively, these data support that modulating the AdvSca1-SM population confers increased protection from the development of unstable atherosclerotic plaques.

4.
JCI Insight ; 8(9)2023 05 08.
Article En | MEDLINE | ID: mdl-36976650

Vascular smooth muscle-derived Sca1+ adventitial progenitor (AdvSca1-SM) cells are tissue-resident, multipotent stem cells that contribute to progression of vascular remodeling and fibrosis. Upon acute vascular injury, AdvSca1-SM cells differentiate into myofibroblasts and are embedded in perivascular collagen and the extracellular matrix. While the phenotypic properties of AdvSca1-SM-derived myofibroblasts have been defined, the underlying epigenetic regulators driving the AdvSca1-SM-to-myofibroblast transition are unclear. We show that the chromatin remodeler Smarca4/Brg1 facilitates AdvSca1-SM myofibroblast differentiation. Brg1 mRNA and protein were upregulated in AdvSca1-SM cells after acute vascular injury, and pharmacological inhibition of Brg1 by the small molecule PFI-3 attenuated perivascular fibrosis and adventitial expansion. TGF-ß1 stimulation of AdvSca1-SM cells in vitro reduced expression of stemness genes while inducing expression of myofibroblast genes that was associated with enhanced contractility; PFI blocked TGF-ß1-induced phenotypic transition. Similarly, genetic knockdown of Brg1 in vivo reduced adventitial remodeling and fibrosis and reversed AdvSca1-SM-to-myofibroblast transition in vitro. Mechanistically, TGF-ß1 promoted redistribution of Brg1 from distal intergenic sites of stemness genes and recruitment to promoter regions of myofibroblast-related genes, which was blocked by PFI-3. These data provide insight into epigenetic regulation of resident vascular progenitor cell differentiation and support that manipulating the AdvSca1-SM phenotype will provide antifibrotic clinical benefits.


Myofibroblasts , Vascular System Injuries , Humans , Myofibroblasts/metabolism , Transforming Growth Factor beta1/metabolism , Chromatin/metabolism , Vascular System Injuries/metabolism , Vascular System Injuries/pathology , Epigenesis, Genetic , Cell Differentiation , Muscle, Smooth, Vascular , Fibrosis , DNA Helicases/genetics , DNA Helicases/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Circulation ; 145(7): 513-530, 2022 02 15.
Article En | MEDLINE | ID: mdl-35000411

BACKGROUND: Aortic valve stenosis is a sexually dimorphic disease, with women often presenting with sustained fibrosis and men with more extensive calcification. However, the intracellular molecular mechanisms that drive these clinically important sex differences remain underexplored. METHODS: Hydrogel biomaterials were designed to recapitulate key aspects of the valve tissue microenvironment and to serve as a culture platform for sex-specific valvular interstitial cells (VICs; precursors to profibrotic myofibroblasts). The hydrogel culture system was used to interrogate intracellular pathways involved in sex-dependent VIC-to-myofibroblast activation and deactivation. RNA sequencing was used to define pathways involved in driving sex-dependent activation. Interventions with small molecule inhibitors and siRNA transfections were performed to provide mechanistic insight into sex-specific cellular responses to microenvironmental cues, including matrix stiffness and exogenously delivered biochemical factors. RESULTS: In both healthy porcine and human aortic valves, female leaflets had higher baseline activation of the myofibroblast marker α-smooth muscle actin compared with male leaflets. When isolated and cultured, female porcine and human VICs had higher levels of basal α-smooth muscle actin stress fibers that further increased in response to the hydrogel matrix stiffness, both of which were higher than in male VICs. A transcriptomic analysis of male and female porcine VICs revealed Rho-associated protein kinase signaling as a potential driver of this sex-dependent myofibroblast activation. Furthermore, we found that genes that escape X-chromosome inactivation such as BMX and STS (encoding for Bmx nonreceptor tyrosine kinase and steroid sulfatase, respectively) partially regulate the elevated female myofibroblast activation through Rho-associated protein kinase signaling. This finding was confirmed by treating male and female VICs with endothelin-1 and plasminogen activator inhibitor-1, factors that are secreted by endothelial cells and known to drive myofibroblast activation through Rho-associated protein kinase signaling. CONCLUSIONS: Together, in vivo and in vitro results confirm sex dependencies in myofibroblast activation pathways and implicate genes that escape X-chromosome inactivation in regulating sex differences in myofibroblast activation and subsequent aortic valve stenosis progression. Our results underscore the importance of considering sex as a biological variable to understand the molecular mechanisms of aortic valve stenosis and to help guide sex-based precision therapies.


Aortic Valve/cytology , Gene Expression , Genes, X-Linked , Myofibroblasts/metabolism , X Chromosome Inactivation , Actins/genetics , Actins/metabolism , Animals , Aortic Valve Stenosis/etiology , Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/pathology , Biomarkers , Cells, Cultured , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Humans , Immunohistochemistry , Male , Myofibroblasts/drug effects , Sex Factors , Signal Transduction , Swine , Transcriptome
7.
Cardiovasc Res ; 118(6): 1452-1465, 2022 05 06.
Article En | MEDLINE | ID: mdl-33989378

Cardiovascular diseases are characterized by chronic vascular dysfunction and provoke pathological remodelling events, such as neointima formation, atherosclerotic lesion development, and adventitial fibrosis. While lineage-tracing studies have shown that phenotypically modulated smooth muscle cells (SMCs) are the major cellular component of neointimal lesions, the cellular origins and microenvironmental signalling mechanisms that underlie remodelling along the adventitial vascular layer are not fully understood. However, a growing body of evidence supports a unique population of adventitial lineage-restricted progenitor cells expressing the stem cell marker, stem cell antigen-1 (Sca1; AdvSca1 cells) as important effectors of adventitial remodelling and suggests that they are at least partially responsible for subsequent pathological changes that occur in the media and intima. AdvSca1 cells are being studied in murine models of atherosclerosis, perivascular fibrosis, and neointima formation in response to acute vascular injury. Depending on the experimental conditions, AdvSca1 cells exhibit the capacity to differentiate into SMCs, endothelial cells, chondrocytes, adipocytes, and pro-remodelling cells, such as myofibroblasts and macrophages. These data indicate that AdvSca1 cells may be a targetable cell population to influence the outcomes of pathologic vascular remodelling. Important questions remain regarding the origins of AdvSca1 cells and the essential signalling mechanisms and microenvironmental factors that regulate both maintenance of their stem-like, progenitor phenotype and their differentiation into lineage-specified cell types. Adding complexity to the story, recent data indicate that the collective population of adventitial progenitor cells is likely composed of several smaller, lineage-restricted subpopulations, which are not fully defined by their transcriptomic profile and differentiation capabilities. The aim of this review is to outline the heterogeneity of Sca1+ adventitial progenitor cells, summarize their role in vascular homeostasis and remodelling, and comment on their translational relevance in humans.


Atherosclerosis , Spinocerebellar Ataxias , Animals , Atherosclerosis/metabolism , Cell Differentiation/genetics , Endothelial Cells/pathology , Fibrosis , Homeostasis , Mice , Myocytes, Smooth Muscle/metabolism , Neointima/metabolism , Spinocerebellar Ataxias/metabolism , Spinocerebellar Ataxias/pathology , Stem Cells/metabolism , Vascular Remodeling
8.
J Am Coll Cardiol ; 78(18): 1782-1795, 2021 11 02.
Article En | MEDLINE | ID: mdl-34711337

BACKGROUND: The effects of nonphysiological flow generated by continuous-flow (CF) left ventricular assist devices (LVADs) on the aorta remain poorly understood. OBJECTIVES: The authors sought to quantify indexes of fibrosis and determine the molecular signature of post-CF-LVAD vascular remodeling. METHODS: Paired aortic tissue was collected at CF-LVAD implant and subsequently at transplant from 22 patients. Aortic wall morphometry and fibrillar collagen content (a measure of fibrosis) was quantified. In addition, whole-transcriptome profiling by RNA sequencing and follow-up immunohistochemistry were performed to evaluate CF-LVAD-mediated changes in aortic mRNA and protein expression. RESULTS: The mean age was 52 ± 12 years, with a mean duration of CF-LVAD of 224 ± 193 days (range 45-798 days). There was a significant increase in the thickness of the collagen-rich adventitial layer from 218 ± 110 µm pre-LVAD to 410 ± 209 µm post-LVAD (P < 0.01). Furthermore, there was an increase in intimal and medial mean fibrillar collagen intensity from 22 ± 11 a.u. pre-LVAD to 41 ± 24 a.u. post-LVAD (P < 0.0001). The magnitude of this increase in fibrosis was greater among patients with longer durations of CF-LVAD support. CF-LVAD led to profound down-regulation in expression of extracellular matrix-degrading enzymes, such as matrix metalloproteinase-19 and ADAMTS4, whereas no evidence of fibroblast activation was noted. CONCLUSIONS: There is aortic remodeling and fibrosis after CF-LVAD that correlates with the duration of support. This fibrosis is due, at least in part, to suppression of extracellular matrix-degrading enzyme expression. Further research is needed to examine the contribution of nonphysiological flow patterns on vascular function and whether modulation of pulsatility may improve vascular remodeling and long-term outcomes.


Aortic Diseases , Assisted Circulation , Extracellular Matrix/enzymology , Heart Failure/therapy , Heart-Assist Devices/adverse effects , ADAMTS4 Protein/metabolism , Aortic Diseases/etiology , Aortic Diseases/pathology , Aortic Diseases/physiopathology , Assisted Circulation/adverse effects , Assisted Circulation/instrumentation , Assisted Circulation/methods , Female , Fibrosis , Humans , Immunohistochemistry , Long Term Adverse Effects/pathology , Male , Matrix Metalloproteinases, Secreted/metabolism , Middle Aged , Sequence Analysis, RNA/methods , Vascular Remodeling/physiology
9.
JCI Insight ; 5(23)2020 12 03.
Article En | MEDLINE | ID: mdl-33119549

Resident vascular adventitial SCA1+ progenitor (AdvSca1) cells are essential in vascular development and injury. However, the heterogeneity of AdvSca1 cells presents a unique challenge in understanding signaling pathways orchestrating their behavior in homeostasis and injury responses. Using smooth muscle cell (SMC) lineage-tracing models, we identified a subpopulation of AdvSca1 cells (AdvSca1-SM) originating from mature SMCs that undergo reprogramming in situ and exhibit a multipotent phenotype. Here we employed lineage tracing and RNA-sequencing to define the signaling pathways regulating SMC-to-AdvSca1-SM cell reprogramming and AdvSca1-SM progenitor cell phenotype. Unbiased hierarchical clustering revealed that genes related to hedgehog/WNT/beta-catenin signaling were significantly enriched in AdvSca1-SM cells, emphasizing the importance of this signaling axis in the reprogramming event. Leveraging AdvSca1-SM-specific expression of GLI-Kruppel family member GLI1 (Gli1), we generated Gli1-CreERT2-ROSA26-YFP reporter mice to selectively track AdvSca1-SM cells. We demonstrated that physiologically relevant vascular injury or AdvSca1-SM cell-specific Kruppel-like factor 4 (Klf4) depletion facilitated the proliferation and differentiation of AdvSca1-SM cells to a profibrotic myofibroblast phenotype rather than macrophages. Surprisingly, AdvSca1-SM cells selectively contributed to adventitial remodeling and fibrosis but little to neointima formation. Together, these findings strongly support therapeutics aimed at preserving the AdvSca1-SM cell phenotype as a viable antifibrotic approach.


Kruppel-Like Transcription Factors/metabolism , Vascular Remodeling/genetics , Animals , Arteries/metabolism , Cell Differentiation/genetics , Female , Fibrosis/genetics , Fibrosis/metabolism , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Myofibroblasts/metabolism , Stem Cells/metabolism , Vascular Remodeling/physiology , Wnt Signaling Pathway
10.
Arterioscler Thromb Vasc Biol ; 40(8): 1854-1869, 2020 08.
Article En | MEDLINE | ID: mdl-32580634

OBJECTIVE: Our recent work demonstrates that PTEN (phosphatase and tensin homolog) is an important regulator of smooth muscle cell (SMC) phenotype. SMC-specific PTEN deletion promotes spontaneous vascular remodeling and PTEN loss correlates with increased atherosclerotic lesion severity in human coronary arteries. In mice, PTEN overexpression reduces plaque area and preserves SMC contractile protein expression in atherosclerosis and blunts Ang II (angiotensin II)-induced pathological vascular remodeling, suggesting that pharmacological PTEN upregulation could be a novel therapeutic approach to treat vascular disease. Approach and Results: To identify novel PTEN activators, we conducted a high-throughput screen using a fluorescence based PTEN promoter-reporter assay. After screening ≈3400 compounds, 11 hit compounds were chosen based on level of activity and mechanism of action. Following in vitro confirmation, we focused on 5-azacytidine, a DNMT1 (DNA methyltransferase-1) inhibitor, for further analysis. In addition to PTEN upregulation, 5-azacytidine treatment increased expression of genes associated with a differentiated SMC phenotype. 5-Azacytidine treatment also maintained contractile gene expression and reduced inflammatory cytokine expression after PDGF (platelet-derived growth factor) stimulation, suggesting 5-azacytidine blocks PDGF-induced SMC de-differentiation. However, these protective effects were lost in PTEN-deficient SMCs. These findings were confirmed in vivo using carotid ligation in SMC-specific PTEN knockout mice treated with 5-azacytidine. In wild type controls, 5-azacytidine reduced neointimal formation and inflammation while maintaining contractile protein expression. In contrast, 5-azacytidine was ineffective in PTEN knockout mice, indicating that the protective effects of 5-azacytidine are mediated through SMC PTEN upregulation. CONCLUSIONS: Our data indicates 5-azacytidine upregulates PTEN expression in SMCs, promoting maintenance of SMC differentiation and reducing pathological vascular remodeling in a PTEN-dependent manner.


DNA (Cytosine-5-)-Methyltransferase 1/antagonists & inhibitors , High-Throughput Screening Assays/methods , PTEN Phosphohydrolase/physiology , Vascular Remodeling/drug effects , Animals , Azacitidine/pharmacology , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/drug effects , PTEN Phosphohydrolase/genetics , Platelet-Derived Growth Factor/pharmacology , Promoter Regions, Genetic
11.
Arterioscler Thromb Vasc Biol ; 40(2): 394-403, 2020 02.
Article En | MEDLINE | ID: mdl-31852223

OBJECTIVE: Pathological vascular remodeling and excessive perivascular fibrosis are major contributors to reduced vessel compliance that exacerbates cardiovascular diseases, for instance, promoting clinically relevant myocardial remodeling. Inflammation plays a significant role in both pathological vascular remodeling and fibrosis. We previously demonstrated that smooth muscle cell-specific PTEN depletion promotes significant vascular fibrosis and accumulation of inflammatory cells. In the current study, we aimed to determine the beneficial role of systemic PTEN elevation on Ang II (angiotensin II)-induced vascular fibrosis and remodeling. Approach and Results: Transgenic mice carrying additional copies of the wild-type Pten gene (super PTEN [sPTEN]) and WT littermates were subjected to Ang II or saline infusion for 14 or 28 days. Compared with WT, Ang II-induced vascular fibrosis was significantly blunted in sPTEN mice, as shown by histochemical stainings and label-free second harmonic generation imaging. The protection against Ang II was recapitulated in sPTEN mice bearing WT bone marrow but not in WT mice reconstituted with sPTEN bone marrow. Ang II-induced elevation of profibrotic and proinflammatory gene expression observed in WT mice was blocked in aortic tissue of sPTEN mice. Immunofluorescent staining and flow cytometry both indicated that perivascular infiltration of T cells and macrophages was significantly inhibited in sPTEN mice. In vitro induction of PTEN expression suppressed Ang II-induced Ccl2 expression in vascular smooth muscle cells. CONCLUSIONS: Systemic PTEN elevation mediates protection against Ang II-induced vascular inflammation and fibrosis predominantly through effects in resident vascular cells. Our data highly support that pharmacological upregulation of PTEN could be a novel and viable approach for the treatment of pathological vascular fibrosis.


Gene Expression Regulation , Muscle, Smooth, Vascular/metabolism , PTEN Phosphohydrolase/genetics , Vascular Diseases/genetics , Vascular Remodeling/genetics , Angiotensin II/toxicity , Animals , Blotting, Western , Cells, Cultured , Disease Models, Animal , Fibrosis/genetics , Fibrosis/metabolism , Fibrosis/pathology , Flow Cytometry , Male , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/pathology , PTEN Phosphohydrolase/biosynthesis , RNA/genetics , Rats , Vascular Diseases/metabolism , Vascular Diseases/pathology
12.
Circ Heart Fail ; 11(5): e004491, 2018 05.
Article En | MEDLINE | ID: mdl-29724722

BACKGROUND: Coronary artery fluid dynamics may be altered because of the nonphysiological flow seen in continuous-flow left ventricular assist devices (CF-LVADs). Our aim was to study the structure and composition of coronary vessels after CF-LVAD. METHODS AND RESULTS: Coronary arteries were collected from patients with heart failure (HF) at the time of transplantation, of whom 15 were supported with a CF-LVAD before transplant (HF+LVAD group) and 9 were not (HF non-LVAD group). In addition, coronary samples were obtained from 5 nonfailing age-matched donors (nonfailing group). Histological analysis was performed to quantify coronary morphology, composition, vascular fibrosis, and vasa vasorum density. The age and sex mix of the 3 groups were similar, and the mean duration of LVAD support was 213 days. Compared with patients with HF and nonfailing donors, the arteries from patients with HF+LVAD had expansion of the adventitia, breakdown of the internal elastic lamina, and increased adventitial collagen deposition and density of vasa vasorum. CONCLUSIONS: Among patients supported with CF-LVADs, the coronary arteries develop marked remodeling with increased adventitial fibrosis. The physiological consequences of these structural changes are unknown, but it is possible that arterial contractility may be impaired, thus limiting coronary flow reserve and promoting myocardial ischemia. This may contribute to CF-LVAD complications, such as ventricular arrhythmias and right ventricular failure. As more patients receive CF-LVADs and new pump technology attempts to modulate flow profiles and pulsatility, further research is needed to understand the mechanisms and long-term sequela of these changes in coronary arteries and other vascular beds.


Coronary Vessels/physiopathology , Fibrosis/physiopathology , Heart Failure/physiopathology , Heart-Assist Devices , Adult , Aged , Female , Heart/physiopathology , Humans , Male , Middle Aged , Myocardial Ischemia/physiopathology , Ventricular Function, Left/physiology
13.
JCI Insight ; 3(4)2018 02 22.
Article En | MEDLINE | ID: mdl-29467331

Phosphatase and tensin homolog (PTEN) is an essential regulator of the differentiated vascular smooth muscle cell (SMC) phenotype. Our goal was to establish that PTEN loss promotes SMC dedifferentiation and pathological vascular remodeling in human atherosclerotic coronary arteries and nonatherosclerotic coronary arteries exposed to continuous-flow left ventricular assist devices (CF-LVADs). Arteries were categorized as nonatherosclerotic hyperplasia (NAH), atherosclerotic hyperplasia (AH), or complex plaque (CP). NAH coronary arteries from CF-LVAD patients were compared to NAH coronaries from non-LVAD patients. Intimal PTEN and SMC contractile protein expression was reduced compared with the media in arteries with NAH, AH, or CP. Compared with NAH, PTEN and SMC contractile protein expression was reduced in the media and intima of arteries with AH and CP. NAH arteries from CF-LVAD patients showed marked vascular remodeling and reduced PTEN and α-smooth muscle actin (αSMA) in medial SMCs compared with arteries from non-LVAD patients; this correlated with increased medial collagen deposition. Mechanistically, compared with ApoE-/- mice, SMC-specific PTEN-null/ApoE-/- double-knockout mice exhibited accelerated atherosclerosis progression and increased vascular fibrosis. By microarray and validated quantitative RT-PCR analysis, SMC PTEN deficiency promotes a global upregulation of proinflammatory and profibrotic genes. We propose that PTEN is an antiinflammatory, antifibrotic target that functions to maintain SMC differentiation. SMC loss of PTEN results in pathological vascular remodeling of human arteries.


Atherosclerosis/pathology , Coronary Vessels/pathology , Heart-Assist Devices/adverse effects , Myocytes, Smooth Muscle/pathology , PTEN Phosphohydrolase/deficiency , Vascular Remodeling , Actins/metabolism , Adult , Aged , Animals , Atherosclerosis/genetics , Cell Differentiation , Coronary Vessels/cytology , Disease Models, Animal , Endothelium, Vascular , Female , Fibrosis , Heart Failure/surgery , Humans , Hyperplasia/pathology , Male , Mice , Mice, Knockout, ApoE , Middle Aged , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/pathology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism
14.
Nat Commun ; 7: 10830, 2016 Mar 04.
Article En | MEDLINE | ID: mdl-26940659

Vascular disease progression is associated with marked changes in vascular smooth muscle cell (SMC) phenotype and function. SMC contractile gene expression and, thus differentiation, is under direct transcriptional control by the transcription factor, serum response factor (SRF); however, the mechanisms dynamically regulating SMC phenotype are not fully defined. Here we report that the lipid and protein phosphatase, PTEN, has a novel role in the nucleus by functioning as an indispensible regulator with SRF to maintain the differentiated SM phenotype. PTEN interacts with the N-terminal domain of SRF and PTEN-SRF interaction promotes SRF binding to essential promoter elements in SM-specific genes. Factors inducing phenotypic switching promote loss of nuclear PTEN through nucleo-cytoplasmic translocation resulting in reduced myogenically active SRF, but enhanced SRF activity on target genes involved in proliferation. Overall decreased expression of PTEN was observed in intimal SMCs of human atherosclerotic lesions underlying the potential clinical importance of these findings.


Myocytes, Smooth Muscle/physiology , PTEN Phosphohydrolase/metabolism , Serum Response Factor/metabolism , Animals , Cell Differentiation , Cells, Cultured , Gene Expression Regulation/physiology , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Muscle, Smooth, Vascular/cytology , PTEN Phosphohydrolase/genetics , Rats , Rats, Sprague-Dawley , Serum Response Factor/genetics
15.
Am J Physiol Renal Physiol ; 301(6): F1326-33, 2011 Dec.
Article En | MEDLINE | ID: mdl-21921021

Diabetic complications of nephropathy and accelerated atherosclerosis are associated with vascular remodeling and dysregulated angiogenesis. Matrix metalloproteinases (MMP) modify extracellular matrix during vascular remodeling and are excreted in urine of patients with vascular malformation or tumor angiogenesis. We hypothesized that urinary MMP activities would be sensitive biomarkers for vascular remodeling in diabetic complications. Activities of MMP-2, MMP-9, and its complex with neutrophil gelatinase-associated lipocalin (NGAL/MMP-9) were measured by substrate gel zymography in urine from nondiabetic (ND) and type 1 diabetic (T1D) rodents that were susceptible to both T1D-induced plaque angiogenesis and nephropathy, or nephropathy alone. Additionally, these urine activities were measured in ND and T1D adolescents. Urinary MMP-9, MMP-2, and NGAL/MMP-9 activities were increased and more prevalent in T1D compared with ND controls. Urinary MMP-2 activity was detected in mice with T1D-induced plaque neovascularization. In nephropathy models, urinary NGAL/MMP-9 and MMP-9 activities appeared before onset of albuminuria, whereas MMP-2 was absent or delayed. Finally, urinary MMP activities were increased in adolescents with early stages of T1D. Urinary MMP activities may be sensitive, noninvasive, and clinically useful biomarkers for predicting vascular remodeling in diabetic renal and vascular complications.


Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Type 1/enzymology , Diabetic Nephropathies/enzymology , Matrix Metalloproteinases/metabolism , Neovascularization, Pathologic/enzymology , Plaque, Atherosclerotic/enzymology , Adolescent , Animals , Biomarkers/urine , Child , Diabetes Mellitus, Experimental/urine , Diabetes Mellitus, Type 1/urine , Female , Humans , Male , Matrix Metalloproteinases/urine , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Young Adult
16.
PLoS One ; 5(11): e13919, 2010 Nov 10.
Article En | MEDLINE | ID: mdl-21085708

BACKGROUND: Lipoprotein lipase (Lpl) acts on triglyceride-rich lipoproteins in the peripheral circulation, liberating free fatty acids for energy metabolism or storage. This essential enzyme is synthesized in parenchymal cells of adipose tissue, heart, and skeletal muscle and migrates to the luminal side of the vascular endothelium where it acts upon circulating lipoproteins. Prior studies suggested that Lpl is immobilized by way of heparan sulfate proteoglycans on the endothelium, but genetically altering endothelial cell heparan sulfate had no effect on Lpl localization or lipolysis. The objective of this study was to determine if extracellular matrix proteoglycans affect Lpl distribution and triglyceride metabolism. METHODS AND FINDINGS: We examined mutant mice defective in collagen XVIII (Col18), a heparan sulfate proteoglycan present in vascular basement membranes. Loss of Col18 reduces plasma levels of Lpl enzyme and activity, which results in mild fasting hypertriglyceridemia and diet-induced hyperchylomicronemia. Humans with Knobloch Syndrome caused by a null mutation in the vascular form of Col18 also present lower than normal plasma Lpl mass and activity and exhibit fasting hypertriglyceridemia. CONCLUSIONS: This is the first report demonstrating that Lpl presentation on the lumenal side of the endothelium depends on a basement membrane proteoglycan and demonstrates a previously unrecognized phenotype in patients lacking Col18.


Basement Membrane/metabolism , Collagen Type XVIII/metabolism , Hypertriglyceridemia/metabolism , Lipoprotein Lipase/metabolism , Animals , Collagen Type XVIII/blood , Collagen Type XVIII/genetics , Encephalocele/blood , Encephalocele/genetics , Encephalocele/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation, Enzymologic , Heparan Sulfate Proteoglycans/genetics , Heparan Sulfate Proteoglycans/metabolism , Humans , Hypertriglyceridemia/genetics , Hypertriglyceridemia/pathology , Immunohistochemistry , Lipoprotein Lipase/blood , Lipoprotein Lipase/genetics , Lipoproteins/blood , Lipoproteins/metabolism , Lipoproteins/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Mutation , Retinal Degeneration , Retinal Detachment/blood , Retinal Detachment/congenital , Retinal Detachment/genetics , Retinal Detachment/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Triglycerides/blood , Triglycerides/metabolism
17.
Arterioscler Thromb Vasc Biol ; 29(11): 1764-71, 2009 Nov.
Article En | MEDLINE | ID: mdl-19729613

OBJECTIVE: Abdominal aortic aneurysm (AAA) is a life-threatening disease affecting almost 10% of the population over age 65. Generation of AAAs by infusion of angiotensin (Ang) II in apolipoprotein E-knockout (ApoE(-/-)) mice is an animal model which supports an imbalance of the renin-angiotensin system in the pathogenesis of AAA. The effect of statins on AngII-mediated AAA formation and the associated neovascularization is not known. Here we determined the effect of simvastatin and the ERK inhibitor, CI1040, on AngII-stimulated AAA formation. METHODS AND RESULTS: ApoE(-/-) mice infused for 28 days with AngII using osmotic minipumps were treated with placebo, 10 mg/kg/d simvastatin, or 100 mg/kg/d CI1040. 95% of AngII-treated mice developed AAA with neovascularization of the lesion, increased ERK phosphorylation, MCP-1 secretion, and MMP activity. These effects were markedly reversed by simvastatin and in part by CI1040. Furthermore, simvastatin and the ERK inhibitor U0126 reversed AngII-stimulated angiogenesis and MMP secretion by human umbilical vein endothelial cells. CONCLUSIONS: These data support the conclusion that simvastatin interferes with AAA formation induced by AngII in ApoE(-/-) mice at least in part via ERK inhibition.


Aortic Aneurysm, Abdominal/prevention & control , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , MAP Kinase Kinase Kinase 3/antagonists & inhibitors , Simvastatin/pharmacology , Angiotensin II , Animals , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Apolipoproteins E/metabolism , Apolipoproteins E/pharmacology , Benzamides/pharmacology , Blood Pressure/drug effects , Blotting, Western , Disease Models, Animal , Immunohistochemistry , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Random Allocation , Reference Values , Renin-Angiotensin System/drug effects
18.
Circ Res ; 101(4): 357-67, 2007 Aug 17.
Article En | MEDLINE | ID: mdl-17615372

Atherosclerosis-related events are a major cause of morbidity and death worldwide, but the mechanisms underlying atherogenesis are not fully understood. We showed in previous studies that the actin-binding protein profilin-1 (pfn) was upregulated in atherosclerotic plaques and in endothelial cells (ECs) treated with oxidized low-density lipoproteins (oxLDL). The present study addressed the role of pfn in atheroma formation. To this end, mice with heterozygous deficiency of pfn, Pfn(+/-), were crossed with Ldlr(-/-) mice. After 2 months under a 1.25% cholesterol atherogenic diet, Pfn(+/-)Ldlr(-/-) (PfnHet) exhibited a significant reduction in lesion burden compared with Ldlr(-/-) control mice (PfnWT), whereas total cholesterol and triglyceride levels were similar in the 2 groups. Relevant atheroprotective changes were identified in PfnHet. When compared with PfnWT, aortas from PfnHet mice showed preserved endothelial nitric oxide synthase (eNOS) activation and nitric oxide (NO)-dependent signaling, and reduced vascular cell adhesion molecule (VCAM)-1 expression and macrophage accumulation at lesion-prone sites. Similarly, knockdown of pfn in cultured aortic ECs was protective against endothelial dysfunction triggered by oxLDL. Finally, bone marrow-derived macrophages from PfnHet showed blunted internalization of oxLDL and oxLDL-induced inflammation. These studies demonstrate that pfn levels modulate processes critical for early atheroma formation and suggest that pfn heterozygosity confers atheroprotection through combined endothelial- and macrophage-dependent mechanisms.


Atherosclerosis/physiopathology , Profilins/genetics , Receptors, LDL/genetics , Vasculitis/physiopathology , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Aorta, Thoracic/physiopathology , Atherosclerosis/metabolism , Atherosclerosis/pathology , CD36 Antigens/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Female , Heterozygote , Lipoproteins, LDL/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III , Profilins/metabolism , Signal Transduction/physiology , Vascular Cell Adhesion Molecule-1/metabolism , Vasculitis/metabolism , Vasculitis/pathology
19.
Curr Opin Lipidol ; 17(5): 548-55, 2006 Oct.
Article En | MEDLINE | ID: mdl-16960504

PURPOSE OF REVIEW: The present review summarizes evidence for several functions of neovascularization in plaque growth that sustain perfusion beyond limits of diffusion from the artery lumen and outer adventitial vasa vasorum, deposit proatherogenic plasma molecules, recruit immune cells and progenitors, and promote intraplaque hemorrhage. Recent approvals of antiangiogenesis drugs for clinical use in cancer and macular degeneration improve the feasibility of testing whether such agents inhibit plaque angiogenesis and incidental atherosclerosis. RECENT FINDINGS: Improvements in large and small animal models of atherosclerosis and knowledge of the molecular regulation of angiogenesis in development and disease have advanced understanding of plaque angiogenesis. Genetic modifications of angiogenesis molecules in mouse strains susceptible to atherosclerosis provide experimental means to identify native molecules that regulate plaque angiogenesis. Studies of plaque angiogenesis are aided by micro-computed tomography techniques that image vasa vasorum anatomy in relation to the atheroma. SUMMARY: Greater knowledge of plaque angiogenesis regulation is needed to design treatments that target the most critical regulatory pathways. Evolutions in angiogenesis inhibitor treatments for cancer and other diseases call for a need to understand the distinct cardiovascular profiles of different agents to rationally combine agents for optimal selectivity and efficacy in the intended vascular bed.


Atherosclerosis/pathology , Neovascularization, Pathologic , Angiogenesis Inhibitors/therapeutic use , Animals , Atherosclerosis/drug therapy , Atherosclerosis/etiology , Atherosclerosis/physiopathology , Collateral Circulation , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Growth Substances/physiology , Humans , Mice , Models, Cardiovascular , Neovascularization, Pathologic/prevention & control
20.
J Lipid Res ; 46(9): 1849-59, 2005 Sep.
Article En | MEDLINE | ID: mdl-15995169

Retention of lipoproteins to proteoglycans in the subendothelial matrix (SEM) is an early event in atherosclerosis. We recently reported that collagen XVIII and its proteolytically released fragment endostatin (ES) are differentially depleted in blood vessels affected by atherosclerosis. Loss of collagen XVIII/ES in atherosclerosis-prone mice enhanced plaque neovascularization and increased the vascular permeability to lipids by distinct mechanisms. Impaired endothelial barrier function increased the influx of lipoproteins across the endothelium; however, we hypothesized that enhanced retention might be a second mechanism leading to the increased lipid content in atheromas lacking collagen XVIII. We now demonstrate a novel property of ES that binds both the matrix proteoglycan biglycan and LDL and interferes with LDL retention to biglycan and to SEM. A peptide encompassing the alpha coil in the ES crystal structure mediates the major blocking effect of ES on LDL retention. ES inhibits the macrophage uptake of biglycan-associated LDL indirectly by interfering with LDL retention to biglycan, but it has no direct effect on the macrophage uptake of native or modified lipoproteins. Thus, loss of ES in advanced atheromas enhances lipoprotein retention in SEM. Our data reveal a third protective role of this vascular basement membrane component during atherosclerosis.


Arteriosclerosis/metabolism , Endostatins/metabolism , Endostatins/pharmacology , Endothelium/metabolism , Lipoproteins, LDL/metabolism , Proteoglycans/metabolism , Animals , Aorta/chemistry , Basement Membrane/metabolism , Biglycan , Binding Sites , Binding, Competitive , Collagen Type XVIII/metabolism , Endostatins/analysis , Extracellular Matrix Proteins , Glycosaminoglycans/metabolism , Humans , Lipoproteins/metabolism , Macrophages/metabolism , Macrophages, Peritoneal , Mice , Mice, Inbred C57BL , Recombinant Proteins , Umbilical Veins
...