Abstract 247: Scaffold-stiffness Dependent Notch1 Activation Regulates Cardiogenic Gene Expression In Cardiac Progenitor Cells

2013 ◽  
Vol 113 (suppl_1) ◽  
Author(s):  
Archana V Boopathy ◽  
Pao L Che ◽  
Yoshie Narui ◽  
Khalid Salaita ◽  
Michael E Davis

Rationale: Cardiac progenitor cells (CPCs) are multipotent, self-renewing cells that can regenerate the myocardium and improve cardiac function in animal models of myocardial infarction (MI). However, limited survival of stem/progenitor cells inhibits cardiac regeneration. Force dependent Notch activation promotes cardiac development and cardiac gene expression in many adult stem cells. As dysregulation of Notch signaling leads to embryonic lethal cardiovascular defects, activating this critical pathway during cell transplantation could improve efficacy of stem cell therapy. Objective: Investigate i) whether self-assembling peptide scaffolds can be used to activate Notch1 signaling in CPCs to promote cardiogenic differentiation and ii) the effect of scaffold stiffness on Notch1 activation and differentiation. Methods: Rat CPCs (c-kit + ) were cultured for 48h in 3D self-assembling scaffolds of varying stiffness (1% low, 2% high): empty scaffolds (RADA), scaffolds modified with peptide mimicking Notch1 ligand, Jagged1 (RJAG), or scaffolds modified with a scrambled peptide (RSCR) and cardiogenic gene expression measured by qRT-PCR. CHO cells expressing Notch1 responsive YFP were also cultured in the above scaffolds for 48h and YFP expression was determined. Results are mean ± SEM with p<0.05 considered significant by one or two-way ANOVA with appropriate post test. Results: In the Notch1 reporter cells, Notch1 activation increased significantly in presence of RJAG (p<0.01) and on increasing scaffold stiffness (p<0.01,n=6) indicating scaffold stiffness-dependent Notch1 activation. Culture of CPCs in RJAG containing 1% scaffolds (low stiffness) significantly increased early endothelial and smooth muscle but not cardiac gene expression while in 2% scaffolds (high stiffness) significantly increased only cardiac and not endothelial or smooth muscle gene expression (p<0.05, n≥4). Conclusions: Taken together, these data show that i) Notch1 activation in 3D is dependent on ligand density and scaffold stiffness and ii) stiffness dependent Notch1 activation differentially regulates cardiogenic gene expression in CPCs. Therefore, delivery of CPCs in JAG containing scaffolds could be used to improve cardiac function following MI.

2012 ◽  
Vol 111 (suppl_1) ◽  
Author(s):  
Archana V Boopathy ◽  
Khalid Salaita ◽  
Michael E Davis

Cardiac progenitor cells (CPCs) are multipotent, self-renewing cells that can regenerate the myocardium and improve cardiac function in animal models of MI by cardiogenic differentiation. However, limited survival of stem/progenitor cells, myocardial scarring and fibrosis inhibit cardiac regeneration. Notch signaling promotes early cardiac development, cardiomyocyte survival and cardiac gene expression in circulating endothelial progenitor cells, mesenchymal stem cells and CPCs. As misregulation of Notch signaling during development is lethal due to cardiovascular defects, activating this critical pathway during cell transplantation could improve the efficacy of stem cell therapy. We investigated whether self-assembling peptide nanofiber hydrogels can be used to activate Notch1 signaling. The 16 amino acid self-assembling scaffold (RAD) was modified with a 20 amino acid peptide mimicking the active site of Notch1 ligand, Jagged1 (RJAG) or with the corresponding scrambled peptide (RSCR). To determine whether scaffold stiffness regulates Notch1 activation, CHO cells with Notch1 responsive YFP expression were cultured in scaffolds of 1-3% w/v in presence of RSCR or RJAG at a 1:10 ligand: scaffold ratio in 3D. Presence of the RJAG peptide (p<0.01) and % concentration of the scaffold (p<0.01) increased Notch1 activation significantly (n=5) indicating that RJAG mediated Notch1 activation in 3D is scaffold stiffness-dependent. Therefore, CPCs were cultured within 3D scaffolds (1-3% w/v; empty, scaffold +RJAG or RSCR) and cardiogenic gene expression was determined by qPCR. An increase in expression of early endothelial (Flk1, Flt1, vWF) and smooth muscle (sm22α, sm αactin) genes was observed in CPCs cultured in 3D scaffolds containing RJAG but not when cultured in 2D. These data show that Notch1 activation is dependent on ligand density and scaffold stiffness. Delivery of CPCs in JAG1 containing self-assembling scaffolds could be used to enhance therapeutic angiogenesis and improve cardiac function following myocardial infarction.


PLoS ONE ◽  
2017 ◽  
Vol 12 (3) ◽  
pp. e0174242 ◽  
Author(s):  
Tareq Al-Maqtari ◽  
Kyung U. Hong ◽  
Bathri N. Vajravelu ◽  
Afsoon Moktar ◽  
Pengxiao Cao ◽  
...  

2012 ◽  
Vol 111 (suppl_1) ◽  
Author(s):  
Karl D Pendergrass ◽  
Michael E Davis

Following acute myocardial infarction, billions of myocytes are lost to cell death. The damage is regional and lost cells are replaced with a collagen scar. One potential therapy to delay or prevent progression into heart failure is regeneration of the damaged myocardium through cell therapy with cardiac progenitor cells (CPC). Reactive oxygen species, specifically hydrogen peroxide, elicit varying responses from different stem/progenitor cells. In the present studies, we sought to determine the effect of acute H2O2 treatment on CPC survival and differentiation. CPCs were isolated and cultured with leukemia inhibitory factor (LIF) to retain their stem-like qualities. CPCs were allowed to differentiate in the absence of LIF for up to 5 days + H2O2. H2O2 (100μM) significantly increased expression of the smooth muscle marker, alpha smooth muscle actin (αSM) by Day 2 as compared to time-matched controls (Ctl: 1.8+1.3 vs 100 μM: 8.5+1.1; p<0.001; N=3). We also observed a trend for an increase in smooth muscle 22 alpha (SM 22α) gene expression by Day 2. Interestingly, by Day 5 the stimulatory effect of 100 μM H2O2 treatment on α SM and SM 22α was reversed and significantly decreased compared to Day 2 (D5: α SM: 0.43+0.05, SM 22α: 0.41+0.2 vs. D2: SM 22α: 10.58+2.3; p<0.01; N=3-4). Evaluation of the endothelial marker VEGFR-2 (Flk-1) showed a trend for an increase in gene expression by Day 2 following 100 μM H2O2 treatment compared to the time-matched control. We also observed an anti-apoptotic effect on CPCs following serum removal, in which 2 days of 100μM H2O2 pretreatment lead to approximately a 55% decrease in cell death compared to untreated CPCs (Ctl: 19.1+2.4 vs 100 μM: 10.5+1.7; p<0.05; N=3-4). The protective effect of the H2O2 pretreatment could be attributed to an increase in anti-oxidative enzymatic capacity in CPCs. There was a trend for an increase in catalase gene expression. In conclusion, our results showed that acute H2O2 preconditioning exerted a stimulatory effect on smooth muscle gene expression and an anti-apoptotic effect compared to time-matched Control CPCs. Furthermore, acute H2O2 preconditioning may aid in directing CPC differentiation towards a vascular phenotype and angiogenesis in the infarcted myocardium, which may prevent or delay heart failure.


2010 ◽  
Vol 49 (6) ◽  
pp. 972-983 ◽  
Author(s):  
Masakuni Tokunaga ◽  
Mei-Lan Liu ◽  
Toshio Nagai ◽  
Koji Iwanaga ◽  
Katsuhisa Matsuura ◽  
...  

2008 ◽  
Vol 51 (23) ◽  
pp. 2289-2290 ◽  
Author(s):  
Stefan Rupp ◽  
Masamichi Koyanagi ◽  
Masayoshi Iwasaki ◽  
Florian Diehl ◽  
Philipp Bushoven ◽  
...  

1998 ◽  
Vol 62 (6) ◽  
pp. 436-442 ◽  
Author(s):  
Naruhito Shimizu ◽  
Minoru Yoshiyama ◽  
Kazuhide Takeuchi ◽  
Akihisa Hanatani ◽  
Shokei Kim ◽  
...  

2008 ◽  
Vol 291 (10) ◽  
pp. 1200-1211 ◽  
Author(s):  
Jian-Fu Chen ◽  
Shusheng Wang ◽  
Qiulian Wu ◽  
Dongsun Cao ◽  
Thiha Nguyen ◽  
...  

2007 ◽  
Vol 309 (2) ◽  
pp. 386
Author(s):  
Allesandro D. Mori ◽  
Yonghong Zhu ◽  
Ilyas Vahora ◽  
Brian Nieman ◽  
Kazuko Koshiba-Takeuchi ◽  
...  

1995 ◽  
Vol 752 (1 Cardiac Growt) ◽  
pp. 370-386 ◽  
Author(s):  
J. L. SAMUEL ◽  
I. DUBUS ◽  
F. FARHADIAN ◽  
F. MAROTTE ◽  
P. OLIVIERO ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document