scholarly journals Functional vascular smooth muscle cells derived from human induced pluripotent stem cells via mesenchymal stem cell intermediates

2012 ◽  
Vol 96 (3) ◽  
pp. 391-400 ◽  
Author(s):  
Vivek K. Bajpai ◽  
Panagiotis Mistriotis ◽  
Yuin-Han Loh ◽  
George Q. Daley ◽  
Stelios T. Andreadis
Stroke ◽  
2021 ◽  
Vol 52 (Suppl_1) ◽  
Author(s):  
Shailaja Rao ◽  
Haruto Uchino ◽  
Arjun V Pendharkar ◽  
Qian Zhang ◽  
Michelle Y Cheng ◽  
...  

Background: Moyamoya disease (MMD) is a rare, progressive steno-occlusive cerebrovascular disorder of the internal carotid artery, leading to stroke. Affected arteries exhibited thickened intima with depleted elastic lamina and media, indicating a dysfunction of the vascular smooth muscle cells (VSMCs) and endothelial cells (ECs). However, the pathogenesis of the disease is still unclear. We aim to address this gap in knowledge by using patient derived induced pluripotent stem cells (iPSCs), to generate VSMCs and ECs. Methods: Peripheral blood mononuclear cells (PBMCs) from controls and MMD patients (n=3 per group) were used for generating iPSCs. Functional properties of differentiated ECs and VSMCs in normoxia/hypoxia model (1%O 2 ) were assessed for cell proliferation by BrDU incorporation, migration by scratch assays and apoptosis by exposure to hydrogen peroxide (H2O2). In vitro angiogenic tube formation was assessed with ECs alone, as well as ECs and VSMCs as a co-culture. Hypoxia inducible factor 1α (HIF1α) and Intercellular adhesion molecule-1 (ICAM-1) activation was determined using qPCR and western blot in VSMCs. Results: Functional proliferative assays showed that MMD ECs proliferated faster than control ECs. Migration assays showed that MMD ECs migrate slower in response to VEGF after hypoxia. MMD ECs were found to be more sensitive to insults such as H2O2 treatment and exhibited more apoptosis. In contrast, MMD VSMCs proliferate and migrate similar to controls, but exhibited elevated levels of HIF1α, ICAM-1 mRNA and protein expression. MMD and control ECs showed similar levels of tube formation in single cultures, however, when co-culturing with VSMCs, MMD VSMCs failed to support EC tubes beyond 24 h, resulting in tube destabilization. Conclusions: Our preliminary results indicate that both MMD VSMCs and ECs are dysfunctional and may be related to the elevated expression of HIF1α and ICAM-1, possibly contributing to MMD pathology. Current ongoing studies include investigating the interactions between MMD VSMCs and ECs using co-cultures, as well as transcriptome analysis of these differentiated cells, which will provide important insights into the cellular and molecular mechanisms underlying MMD.


2021 ◽  
Vol 128 (5) ◽  
pp. 670-686
Author(s):  
Mengcheng Shen ◽  
Thomas Quertermous ◽  
Michael P. Fischbein ◽  
Joseph C. Wu

The developmental origin of vascular smooth muscle cells (VSMCs) has been increasingly recognized as a major determinant for regional susceptibility or resistance to vascular diseases. As a human material-based complement to animal models and human primary cultures, patient induced pluripotent stem cell iPSC-derived VSMCs have been leveraged to conduct basic research and develop therapeutic applications in vascular diseases. However, iPSC-VSMCs (induced pluripotent stem cell VSMCs) derived by most existing induction protocols are heterogeneous in developmental origins. In this review, we summarize signaling networks that govern in vivo cell fate decisions and in vitro derivation of distinct VSMC progenitors, as well as key regulators that terminally specify lineage-specific VSMCs. We then highlight the significance of leveraging patient-derived iPSC-VSMCs for vascular disease modeling, drug discovery, and vascular tissue engineering and discuss several obstacles that need to be circumvented to fully unleash the potential of induced pluripotent stem cells for precision vascular medicine.


2021 ◽  
Vol 129 (Suppl_1) ◽  
Author(s):  
LU LIU ◽  
Adrien Georges ◽  
Nabila Bouatia-Naji

Introduction: The low-density lipoprotein receptor-related protein 1 (LRP1), an endocytic receptor highly expressed in smooth muscle cells (SMCs), participates in diverse biological processes. A common genetic variant located in LRP1 first intron, rs11172113, was associated with several vascular diseases, including coronary artery disease, migraine and spontaneous coronary artery dissection, as well asd with LRP1 expression in arterial tissues. However, the biological mechanisms through which rs11172113 influence LRP1 function in the context of arterial lesions is not fully understood. Methods: We applied in silico functional annotation to select variants and measured their enhancer activity using luciferase reporter assay in rat primary cells (A7r5). We performed siRNA knockdown of LRP1 and 4 transcription factors (TFs) predicted to interact with rs11172113 in human induced pluripotent stem cells (iPSCs) derived SMCs. We analyzed both contractile (CSMCs) and synthetic (SSMCs) differentiated cells. We edited iPSCs prior to differentiation using CRISPR-Cas9 to generate 100 bp deletion of the enhancer region containing rs11172113. We also created frame-shift indels in exons 2 or 5 of LRP1 in iPSCs to create SMCs knockouts. Results: Seven variants in LRP1 locus co-located with enhancer (histone marks) and open chromatin regions (ATAC-Seq peaks) in SMCs and arterial tissues. Reporter assay in rat SMCs confirmed that rs11172113 belongs to a genomic region showing enhancer activity in vitro . iPSCs with homozygous deletion of rs11172113 enhancer region presented the same pluripotency compared with wild type, and iPSC derived SMCs showed positive expression of specific markers for each phenotype. We found that the deletion of enhancer region decreased the expression of LRP1 in both CSMCs and SSMCs. LRP1 knockdown decreased SSMCs and CSMCs proliferation capacity, but increased cell migration. Knockdown of TFs and iPSCs derived CSMCs and SSMCs with LRP1 knockout are currently under assessment. Conclusions: We confirmed rs11172113 to regulate LRP1 expression in iPSCs derived synthetic and contractile SMCs. Our results support LRP1 effect on SMCs phenotype alteration as a potential mechanism in genetic susceptibility for vascular disease.


Sign in / Sign up

Export Citation Format

Share Document