Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Kidney Int ; 95(4): 958-972, 2019 04.
Article in English | MEDLINE | ID: mdl-30827513

ABSTRACT

Children on dialysis have a cardiovascular mortality risk equivalent to older adults in the general population, and rapidly develop medial vascular calcification, an age-associated pathology. We hypothesized that premature vascular ageing contributes to calcification in children with advanced chronic kidney disease (CKD). Vessels from children with Stage 5 CKD with and without dialysis had evidence of increased oxidative DNA damage. The senescence markers p16 and p21 were also increased in vessels from children on dialysis. Treatment of vessel rings ex vivo with calcifying media increased oxidative DNA damage in vessels from children with Stage 5 CKD, but not in those from healthy controls. Vascular smooth muscle cells cultured from children on dialysis exhibited persistent DNA damage, impaired DNA damage repair, and accelerated senescence. Under calcifying conditions vascular smooth muscle cells from children on dialysis showed increased osteogenic differentiation and calcification. These changes correlated with activation of the senescence-associated secretory phenotype (SASP), an inflammatory phenotype characterized by the secretion of proinflammatory cytokines and growth factors. Blockade of ataxia-telangiectasia mutated (ATM)-mediated DNA damage signaling reduced both inflammation and calcification. Clinically, children on dialysis had elevated circulating levels of osteogenic SASP factors that correlated with increased vascular stiffness and coronary artery calcification. These data imply that dysregulated mineral metabolism drives vascular "inflammaging" by promoting oxidative DNA damage, premature senescence, and activation of a pro-inflammatory SASP. Drugs that target DNA damage signaling or eliminate senescent cells may have the potential to prevent vascular calcification in patients with advanced CKD.


Subject(s)
Arteritis/etiology , Cellular Senescence/genetics , Kidney Failure, Chronic/therapy , Renal Dialysis/adverse effects , Vascular Calcification/etiology , Adolescent , Arteries/cytology , Arteries/diagnostic imaging , Arteries/pathology , Arteritis/pathology , Ataxia Telangiectasia Mutated Proteins/metabolism , Cells, Cultured , Child , Child, Preschool , DNA Damage , Female , Humans , Infant , Kidney Failure, Chronic/complications , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Oxidative Stress , Primary Cell Culture , Vascular Calcification/pathology
2.
Sci Transl Med ; 6(227): 227ra34, 2014 03 12.
Article in English | MEDLINE | ID: mdl-24622514

ABSTRACT

Veins grafted into an arterial environment undergo a complex vascular remodeling process. Pathologic vascular remodeling often results in stenosed or occluded conduit grafts. Understanding this complex process is important for improving the outcome of patients with coronary and peripheral artery disease undergoing surgical revascularization. Using in vivo murine cell lineage-tracing models, we show that endothelial-derived cells contribute to neointimal formation through endothelial-to-mesenchymal transition (EndMT), which is dependent on early activation of the Smad2/3-Slug signaling pathway. Antagonism of transforming growth factor-ß (TGF-ß) signaling by TGF-ß neutralizing antibody, short hairpin RNA-mediated Smad3 or Smad2 knockdown, Smad3 haploinsufficiency, or endothelial cell-specific Smad2 deletion resulted in decreased EndMT and less neointimal formation compared to controls. Histological examination of postmortem human vein graft tissue corroborated the changes observed in our mouse vein graft model, suggesting that EndMT is operative during human vein graft remodeling. These data establish that EndMT is an important mechanism underlying neointimal formation in interpositional vein grafts, and identifies the TGF-ß-Smad2/3-Slug signaling pathway as a potential therapeutic target to prevent clinical vein graft stenosis.


Subject(s)
Cell Transdifferentiation/drug effects , Endothelial Cells/drug effects , Mesoderm/drug effects , Signal Transduction , Transforming Growth Factor beta/metabolism , Veins/growth & development , Veins/transplantation , Animals , Antibodies, Neutralizing/pharmacology , Cell Lineage/drug effects , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Knockdown Techniques , Humans , Mesoderm/cytology , Mesoderm/metabolism , Mice , Neointima/metabolism , Signal Transduction/drug effects , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Snail Family Transcription Factors , Transcription Factors/metabolism , Veins/drug effects
3.
Circ Res ; 112(10): e99-109, 2013 May 10.
Article in English | MEDLINE | ID: mdl-23564641

ABSTRACT

RATIONALE: Vascular calcification is prevalent in the aging population, yet little is known of the mechanisms driving age-associated vascular smooth muscle cell (VSMC) phenotypic change. OBJECTIVE: To investigate the role of nuclear lamina disruption, a specific hallmark of VSMC aging, in driving VSMC osteogenic differentiation. METHODS AND RESULTS: Prelamin A, the unprocessed form of the nuclear lamina protein lamin A, accumulated in calcifying human VSMCs in vitro and in vivo, and its overexpression promoted VSMC osteogenic differentiation and mineralization. During VSMC aging in vitro, prelamin A accumulation occurred concomitantly with increased p16 expression and osteogenic differentiation and was associated with increased levels of DNA damage. Microarray analysis showed that DNA damage repair pathways were significantly impaired in VSMCs expressing prelamin A and that chemical inhibition and siRNA depletion of the DNA damage response kinases ataxia-telangiectasia mutated/ataxia-telangiectasia- and Rad3-related effectively blocked VSMC osteogenic differentiation and mineralization. In coculture experiments, prelamin A-expressing VSMCs induced alkaline phosphatase activity in mesenchymal progenitor cells, and this was abrogated by inhibition of ataxia-telangiectasia-mutated signaling, suggesting that DNA damage induces the secretion of pro-osteogenic factors by VSMCs. Cytokine array analysis identified several ataxia-telangiectasia mutated-dependent senescence-associated secretory phenotype factors/cytokines released by prelamin A-positive VSMCs, including the calcification regulators bone morphogenetic protein 2, osteoprotegerin, and interleukin 6. CONCLUSIONS: Prelamin A promotes VSMC calcification and aging by inducing persistent DNA damage signaling, which acts upstream of VSMC osteogenic differentiation and the senescence-associated secretory phenotype. Agents that target the DNA damage response and prelamin A toxicity may be potential therapies for the treatment of vascular calcification.


Subject(s)
Aging/physiology , DNA Damage/physiology , Muscle, Smooth, Vascular/physiopathology , Nuclear Proteins/physiology , Phenotype , Protein Precursors/physiology , Vascular Calcification/physiopathology , Adult , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16 , Cytokines/metabolism , Female , Humans , In Vitro Techniques , Lamin Type A , Male , Middle Aged , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Neoplasm Proteins/physiology , Nuclear Proteins/genetics , Protein Precursors/genetics , Signal Transduction/physiology , Transfection
4.
Stem Cells ; 28(9): 1465-75, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20629173

ABSTRACT

Donor-recipient cell interactions are essential for functional engraftment after nonautologous cell transplantation. During this process, transplant engraftment is characterized and defined by interactions between transplanted cells with local and recruited inflammatory cells. The outcome of these interactions determines donor cell fate. Here, we provide evidence that lineage-committed embryonic stem cell (ESC)-derived vascular progenitor cells are the target of major histocompatibility complex (MHC) class I-dependent, natural killer (NK) cell-mediated elimination in vitro and in vivo. Treatment with interferon γ was found to significantly upregulate MHC class I expression on ESC-derived vascular progenitor cells, rendering them less susceptible to syngeneic NK cell-mediated killing in vitro and enhancing their survival and differentiation potential in vivo. Furthermore, in vivo ablation of NK cells led to enhanced progenitor cell survival after transplantation into a syngeneic murine ischemic hindlimb model, providing additional evidence that NK cells mediate ESC-derived progenitor cell transplant rejection. These data highlight the importance of recipient immune-donor cell interactions, and indicate a functional role for MHC-I antigen expression during successful ESC-derived syngeneic transplant engraftment.


Subject(s)
Embryonic Stem Cells/transplantation , Endothelial Cells/transplantation , Graft Rejection/prevention & control , Graft Survival , Hemangioblasts/transplantation , Histocompatibility Antigens Class I/immunology , Ischemia/surgery , Killer Cells, Natural/immunology , Muscle, Skeletal/blood supply , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Lineage , Cell Survival , Cells, Cultured , Disease Models, Animal , Embryonic Stem Cells/immunology , Embryonic Stem Cells/metabolism , Endothelial Cells/immunology , Endothelial Cells/metabolism , Female , Graft Rejection/immunology , Hemangioblasts/immunology , Hemangioblasts/metabolism , Hindlimb , Interferon-gamma/immunology , Ischemia/immunology , Ischemia/physiopathology , Mice , Mice, Inbred NOD , Mice, SCID , Neovascularization, Physiologic , Time Factors , Transplantation, Isogeneic
5.
J Clin Invest ; 120(1): 303-14, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20038813

ABSTRACT

Inflammation is a key component of arterial injury, with VSMC proliferation and neointimal formation serving as the final outcomes of this process. However, the acute events transpiring immediately after arterial injury that establish the blueprint for this inflammatory program are largely unknown. We therefore studied these events in mice and found that immediately following arterial injury, medial VSMCs upregulated Rantes in an acute manner dependent on Stat3 and NF-kappaB (p65 subunit). This led to early T cell and macrophage recruitment, processes also under the regulation of the cyclin-dependent kinase inhibitor p21Cip1. Unique to VSMCs, Rantes production was initiated by Tnf-alpha, but not by Il-6/gp130. This Rantes production was dependent on the binding of a p65/Stat3 complex to NF-kappaB-binding sites within the Rantes promoter, with shRNA knockdown of either Stat3 or p65 markedly attenuating Rantes production. In vivo, acute NF-kappaB and Stat3 activation in medial VSMCs was identified, with acute Rantes production after injury substantially reduced in Tnfa-/- mice compared with controls. Finally, we generated mice with SMC-specific conditional Stat3 deficiency and confirmed the Stat3 dependence of acute Rantes production by VSMCs. Together, these observations unify inflammatory events after vascular injury, demonstrating that VSMCs orchestrate the arterial inflammatory response program via acute Rantes production and subsequent inflammatory cell recruitment.


Subject(s)
Chemokine CCL5/biosynthesis , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , STAT3 Transcription Factor/physiology , Vasculitis/etiology , Animals , Arteries/metabolism , Cells, Cultured , Chemokine CCL2/biosynthesis , Chemokine CCL5/analysis , Chemokine CCL5/genetics , Cyclin-Dependent Kinase Inhibitor p21/analysis , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/physiology , Female , Interleukin-6/pharmacology , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/physiology , STAT3 Transcription Factor/analysis , STAT3 Transcription Factor/genetics , T-Lymphocytes/physiology , Tumor Necrosis Factor-alpha/pharmacology
6.
J Mol Med (Berl) ; 86(11): 1221-32, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18690419

ABSTRACT

Recruitment and retention of circulating progenitor cells at the site of injured or ischemic tissues facilitates adult neo-vascularization. We hypothesized that cell therapy could modulate local neo-vascularization through the vascular endothelial growth factor (VEGF)/stromal cell-derived factor-1 (SDF-1) axis and by paracrine effects on local endothelial cells. We isolated from rat bone marrow a subset of multipotent adult progenitor cell-derived progenitor cells (MDPC). In vitro, MDPCs secreted multiple cytokines related to inflammation and angiogenesis, including monocyte chemotactic protein-1, SDF-1, basic fibroblast growth factor, and VEGF, and expressed the chemokine receptors CXCR4 and VEGFR1. To investigate in vivo properties, we transplanted MDPCs into the ischemic hind limbs of rats. Elevated levels of the chemokine SDF-1 and colocalization of CD11b(+) cells marked the initial phase of tissue remodeling after cell transplantation. Prolonged engraftment was observed in the adventitial-medial border region of arterioles of ischemic muscles. However, engrafted cells did not differentiate into endothelial or smooth muscle cells. Limb perfusion normalized 4 weeks after cell injection. Inhibition of SDF-1 reduced the engraftment of transplanted cells and decreased endothelial cell proliferation. These findings suggest a two-stage model whereby transplanted MDPCs modulate wound repair through recruitment of inflammatory cells to ischemic tissue. This is an important potential mechanism for cell transplantation, in addition to the direct modulation of local vascular cells through paracrine mechanisms.


Subject(s)
Adult Stem Cells/physiology , Chemokine CXCL12/physiology , Multipotent Stem Cells/physiology , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Receptors, CXCR4/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Adult Stem Cells/pathology , Adult Stem Cells/transplantation , Animals , Bone Marrow Cells/pathology , Bone Marrow Cells/physiology , CD11b Antigen/metabolism , Cell Differentiation , Cell Lineage , Cell Proliferation , Cells, Cultured , Chemokine CXCL12/antagonists & inhibitors , Female , Hindlimb , Inflammation/immunology , Inflammation/pathology , Inflammation/therapy , Ischemia/immunology , Ischemia/pathology , Ischemia/therapy , Microvessels/physiopathology , Multipotent Stem Cells/pathology , Multipotent Stem Cells/transplantation , Muscle, Skeletal/immunology , Paracrine Communication , Rats , Rats, Inbred F344
7.
J Clin Invest ; 118(6): 2050-61, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18464929

ABSTRACT

Cyclin-dependent kinase inhibitors, including p21Cip1, are implicated in cell turnover and are active players in cardiovascular wound repair. Here, we show that p21Cip1 orchestrates the complex interactions between local vascular and circulating immune cells during vascular wound repair. In response to femoral artery mechanical injury, mice with homozygous deletion of p21Cip1 displayed accelerated proliferation of VSMCs and increased immune cell infiltration. BM transplantation experiments indicated that local p21Cip1 plays a pivotal role in restraining excessive proliferation during vascular wound repair. Increased local vascular stromal cell-derived factor-1 (SDF-1) levels were observed after femoral artery injury in p21+/+ and p21-/- mice, although this was significantly greater in p21-/- animals. In addition, disruption of SDF-1/CXCR4 signaling inhibited the proliferative response during vascular remodeling in both p21+/+ and p21-/- mice. We provide evidence that the JAK/STAT signaling pathway is an important regulator of vascular SDF-1 levels and that p21Cip1 inhibits STAT3 binding to the STAT-binding site within the murine SDF-1 promoter. Collectively, these results suggest that p21Cip1 activity is essential for the regulation of cell proliferation and inflammation after arterial injury in local vascular cells and that the SDF-1/CXCR4 signaling system is a key mediator of vascular proliferation in response to injury.


Subject(s)
Apoptosis , Chemokine CXCL12/metabolism , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Receptors, CXCR4/metabolism , Wound Healing , Animals , Cell Proliferation , Femoral Artery/pathology , Homozygote , Mice , Mice, Transgenic , Models, Biological , T-Lymphocytes/metabolism , Time Factors , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...