Schmuck EG et al. (MAR 2014)
Cardiovascular engineering and technology 5 1 119--131
Cardiac fibroblast-derived 3D extracellular matrix seeded with mesenchymal stem cells as a novel device to transfer cells to the ischemic myocardium.
PURPOSE Demonstrate a novel manufacturing method to generate extracellular matrix scaffolds from cardiac fibroblasts (CF-ECM) as a therapeutic mesenchymal stem cell-transfer device. MATERIALS AND METHODS Rat CF were cultured at high-density (˜1.6×10(5)/cm(2)) for 10-14 days. Cell sheets were removed from the culture dish by incubation with EDTA and decellularized with water and peracetic acid. CF-ECM was characterized by mass spectrometry,immunofluorescence and scanning electron microscopy. CF-ECM seeded with human embryonic stem cell derived mesenchymal stromal cells (hEMSCs) were transferred into a mouse myocardial infarction model. 48 hours later,mouse hearts were excised and examined for CF-ECM scaffold retention and cell transfer. RESULTS CF-ECM scaffolds are composed of fibronectin (82%),collagens type I (13%),type III (3.4%),type V (0.2%),type II (0.1%) elastin (1.3%) and 18 non-structural bioactive molecules. Scaffolds remained intact on the mouse heart for 48 hours without the use of sutures or glue. Identified hEMSCs were distributed from the epicardium to the endocardium. CONCLUSIONS High density cardiac fibroblast culture can be used to generate CF-ECM scaffolds. CF-ECM scaffolds seeded with hEMSCs can be maintained on the heart without suture or glue. hEMSC are successfully delivered throughout the myocardium.
View Publication
(Jun 2025)
iScience 28 8
Cardiac fibrosis inhibitor CTPR390 prevents structural and morphological changes in human engineered cardiac connective tissue
SummaryCardiac fibrosis is a key characteristic of heart failure. CTPR390,an experimental anti-fibrotic inhibitor targeting Hsp90,has shown success in animal models,but remains unexplored in human cardiac models. This study evaluated an engineered cardiac connective tissue (ECCT) model,focusing on changes in the extracellular matrix and fibroblasts. Results showed that CTPR390 prevented architectural changes in TGF?1-activated ECCT,preserving tissue perimeter,collagen fibers alignment while reducing structured areas and degree of collagen structuration. CTPR390 treatment reduced cell area of fibroblasts under tension,without changes in the internal rounded cells devoid of tension. Fibroblast recruitment to tension areas was diminished,showing biomechanical behavior similar to control ECCT. This treatment also lowered the gene and protein expression of key pro-fibrotic markers. Here,advanced biotechnology was employed to detect the detailed structure of tissue fibrosis reduction after administering CTPR390,representing a significant advancement toward clinical application for cardiac fibrosis treatment. Graphical abstract Highlights•CTPR390 prevented architectural changes in TGF?1-activated ECCT•CTPR390 preserves tissue perimeter,collagen fibers alignment•CTPR390 reduces structured areas and degree of collagen structuration•CTPR390-trested ECCTs presented a biomechanical behavior similar to control ECCT Molecular biology; Cell biology
View Publication
Wang H et al. (MAR 2011)
ACS chemical biology 6 2 192--7
Cardiac induction of embryonic stem cells by a small molecule inhibitor of Wnt/β-catenin signaling.
In vitro differentiation of embryonic stem cells is tightly regulated by the same key signaling pathways that control pattern formation during embryogenesis. Small molecules that selectively target these developmental pathways,including Wnt,and BMP signaling may be valuable for directing differentiation of pluripotent stem cells toward many desired tissue types,but to date only few such compounds have been shown to promote cardiac differentiation. Here,we show that XAV939,a recently discovered small molecule inhibitor of Wnt/β-catenin signaling,can robustly induce cardiomyogenesis in mouse ES cells. Our results suggest that a timely administration of XAV939 immediately following the formation of mesoderm progenitor cells promotes cardiomyogenic development at the expense of other mesoderm derived lineages,including the endothelial,smooth muscle,and hematopoietic lineages. Given the critical role that Wnt/β-catenin signaling plays in many aspects of embryogenesis and tissue regeneration,XAV939 is a valuable chemical probe to dissect in vitro differentiation of stem cells and to explore their regenerative potential in a variety of contexts.
View Publication
Sadek H et al. ( 2008)
Proceedings of the National Academy of Sciences of the United States of America 105 16 6063--6068
Cardiogenic small molecules that enhance myocardial repair by stem cells.
The clinical success of stem cell therapy for myocardial repair hinges on a better understanding of cardiac fate mechanisms. We have identified small molecules involved in cardiac fate by screening a chemical library for activators of the signature gene Nkx2.5,using a luciferase knockin bacterial artificial chromosome (BAC) in mouse P19CL6 pluripotent stem cells. We describe a family of sulfonyl-hydrazone (Shz) small molecules that can trigger cardiac mRNA and protein expression in a variety of embryonic and adult stem/progenitor cells,including human mobilized peripheral blood mononuclear cells (M-PBMCs). Small-molecule-enhanced M-PBMCs engrafted into the rat heart in proximity to an experimental injury improved cardiac function better than control cells. Recovery of cardiac function correlated with persistence of viable human cells,expressing human-specific cardiac mRNAs and proteins. Shz small molecules are promising starting points for drugs to promote myocardial repair/regeneration by activating cardiac differentiation in M-PBMCs.
View Publication
Yau WW et al. (JAN 2011)
Proteome science 9 1 3
Cardiogenol C can induce Mouse Hair Bulge Progenitor Cells to Transdifferentiate into Cardiomyocyte-like Cells.
BACKGROUND: Hair bulge progenitor cells (HBPCs) are multipotent stem cells derived from the bulge region of mice vibrissal hairs. The purified HBPCs express CD34,K15 and K14 surface markers. It has been reported that HBPCs could be readily induced to transdifferentiate into adipocytes and osteocytes. However,the ability of HBPCs to transdifferentiate into cardiomyocytes has not yet been investigated. METHODOLOGY/PRINCIPAL FINDINGS: The cardiomyogenic potential of HBPCs was investigated using a small cell-permeable molecule called Cardiogenol C. We established that Cardiogenol C could induce HBPCs to express transcription factors GATA4,Nkx2.5 and Tbx5,which are early specific markers for pre-cardiomyogenic cells. In prolonged cultures,the Cardiogenol C-treated HBPCs can also express muscle proteins,cardiac-specific troponin I and sarcomeric myosin heavy chain. However,we did not observe the ability of these cells to functionally contract. Hence,we called these cells cardiomyocyte-like cells rather than cardiomyocytes. We tried to remedy this deficiency by pre-treating HBPCs with Valproic acid first before exposing them to Cardiogenol C. This pretreatment inhibited,rather than improved,the effectiveness of Cardiogenol C in reprogramming the HBPCs. We used comparative proteomics to determine how Cardiogenol C worked by identifying proteins that were differentially expressed. We identified proteins that were involved in promoting cell differentiation,cardiomyocyte development and for the normal function of striated muscles. From those differentially expressed proteins,we further propose that Cardiogenol C might exert its effect by activating the Wnt signaling pathway through the suppression of Kremen1. In addition,by up-regulating the expression of chromatin remodeling proteins,SIK1 and Smarce1 would initiate cardiac differentiation. CONCLUSIONS/SIGNIFICANCE: In conclusion,our CD34+/K15+ HBPCs could be induced to transdifferentiate into cardiomyocyte-like cells using a small molecule called Cardiogenol C. The process involves activation of the Wnt signaling pathway and altered expression of several key chromatin remodeling proteins. The finding is clinically significant as HBPCs offer a readily accessible and autologous source of progenitor cells for cell-based therapy of heart disease,which is one of major killers in developed countries.
View Publication
A. A. Salybekov et al. (Nov 2024)
International Journal of Molecular Sciences 25 21
Cardioimmunology in Health and Diseases: Impairment of the Cardio-Spleno-Bone Marrow Axis Following Myocardial Infarction in Diabetes Mellitus
A comprehensive understanding of the cardio-spleen-bone marrow immune cell axis is essential for elucidating the alterations occurring during the pathogenesis of diabetes mellitus (DM). This study investigates the dynamics of immune cell kinetics in DM after myocardial infarction (MI) over time. MI was induced in diabetic and healthy control groups using C57BL/N6 mice,with sacrifices occurring at days 1,3,7,and 28 post-MI to collect heart,peripheral blood (PB),spleen,and bone marrow (BM) samples. Cell suspensions from each organ were isolated and analyzed via flow cytometry. Additionally,the endothelial progenitor cell-colony-forming assay (EPC-CFA) was performed using mononuclear cells derived from BM,PB,and the spleen. The results indicated that,despite normal production in BM and the spleen,CD45+ cells were lower in the PB of DM mice at days 1 to 3. Further analysis revealed a reduction in total and pro-inflammatory neutrophils (N1s) in PB at days 1 to 3 and in the spleen at days 3 to 7 in DM mice,suggesting that DM-induced alterations in splenic neutrophils fail to meet the demand in PB and ischemic tissues. Infiltrating macrophages (total,M1,M2) were reduced at day 3 in the DM-ischemic heart,with total and M1 (days 1–3) and M2 (days 3–7) macrophages being significantly decreased in DM-PB compared to controls,indicating impaired macrophage recruitment and polarization in DM. Myeloid dendritic cells (mDCs) in the heart were higher from days 1 to 7,which corresponded with the enhanced recruitment of CD8+ cells from days 1 to 28 in the DM-infarcted myocardium. Total CD4+ cells decreased in DM-PB at days 1 to 3,suggesting a delayed adaptive immune response to MI. B cells were reduced in PB at days 1 to 3,in myocardium at day 3,and in the spleen at day 7,indicating compromised mobilization from BM. EPC-CFA results showed a marked decrease in definitive EPC colonies in the spleen and BM from days 1 to 28 in DM mice compared to controls in vitro,highlighting that DM severely impairs EPC colony-forming activity by limiting the differentiation of EPCs from primitive to definitive forms. Taking together,this study underscores significant disruptions in the cardio-spleen-bone marrow immune cell axis following MI in DM,revealing delayed innate and adaptive immune responses along with impaired EPC differentiation.
View Publication
(Jul 2025)
Cell Reports Medicine 6 7
Cardiolipin-mimic lipid nanoparticles without antibody modification delivered senolytic in vivo CAR-T therapy for inflamm-aging
SummarymRNA-based in vivo chimeric antigen receptor (CAR)-T cell engineering offers advantages over ex vivo therapies,including streamlined manufacturing and transient expression. However,current delivery methods require antibody-modified vehicles with manufacturing challenges. In this study,inspired by cardiolipin,we identify cardiolipin-like di-phosphoramide lipids that improve T cell transfection without targeting ligands,both in vitro and in vivo. The T cell-favored tropism is likely due to the lipid’s packing,shape,and rigidity. Encapsulating circular RNA further prolongs mRNA expression in the spleen and T cells. Using PL40 lipid nanoparticles,we deliver mRNA encoding a CAR targeting the senolytic and inflammatory antigen urokinase-type plasminogen activator receptor (uPAR),alleviating uPAR-related liver fibrosis and rheumatoid arthritis (RA). Single-cell sequencing in humans confirms uPAR’s relevance to senescence and inflammation in RA. To facilitate clinical translation,we screen and humanize single-chain variable fragments (scFvs) against uPAR,establishing a PL40 mRNA-encoded humanized uPAR CAR with potential for treating aging-inflamed disorders. Graphical abstract Highlights•Cardiolipin-mimic phosphoramide (CAMP) LNPs transfect T cells without antibody modification•Circular mRNA prolongs mRNA expression•Senolytic in vivo CAR-T treats inflamm-aging disease (liver fibrosis and rheumatoid arthritis)•Develop humanized anti-human uPAR scFv Zhang et al. develop Cardiolipin-mimic phosphoramide (CAMP) lipids,which enable T cell transfection without antibody modification. Using CAMP-based LNPs,they generate senolytic CAR-T cells in vivo to target inflamm-aging diseases. Additionally,they employ circular mRNA to prolong transgene expression. The authors also engineer a humanized anti-human uPAR scFv for clinically relevant applications.
View Publication
Dambrot C et al. (FEB 2011)
The Biochemical journal 434 1 25--35
Cardiomyocyte differentiation of pluripotent stem cells and their use as cardiac disease models.
More than 10 years after their first isolation,human embryonic stem cells are finally 'coming of age' in research and biotechnology applications as protocols for their differentiation and undifferentiated expansion in culture become robust and scalable,and validated commercial reagents become available. Production of human cardiomyocytes is now feasible on a daily basis for many laboratories with tissue culture expertise. An additional recent surge of interest resulting from the first production of human iPSCs (induced pluripotent stem cells) from somatic cells of patients now makes these technologies of even greater importance since it is likely that (genetic) cardiac disease phenotypes can be captured in the cardiac derivatives of these cells. Although cell therapy based on replacing cardiomyocytes lost or dysfunctional owing to cardiac disease are probably as far away as ever,biotechnology and pharmaceutical applications in safety pharmacology and drug discovery will probably impact this clinical area in the very near future. In the present paper,we review the cutting edge of this exciting area of translational research.
View Publication
(Feb 2024)
iScience 27 3
Cardiomyocyte-fibroblast interaction regulates ferroptosis and fibrosis after myocardial injury
SummaryNeonatal mouse hearts have transient renewal capacity,which is lost in juvenile and adult stages. In neonatal mouse hearts,myocardial infarction (MI) causes an initial loss of cardiomyocytes. However,it is unclear which type of regulated cell death (RCD) occurs in stressed cardiomyocytes. In the current studies,we induced MI in neonatal and juvenile mouse hearts and showed that ischemic cardiomyocytes primarily undergo ferroptosis,a non-apoptotic and iron-dependent form of RCD. We demonstrated that cardiac fibroblasts (CFs) protect cardiomyocytes from ferroptosis through paracrine effects and direct cell-cell interaction. CFs show strong resistance to ferroptosis due to high ferritin expression. The fibrogenic activity of CFs,typically considered detrimental to heart function,is negatively regulated by paired-like homeodomain 2 (Pitx2) signaling from cardiomyocytes. In addition,Pitx2 prevents ferroptosis in cardiomyocytes by regulating ferroptotic genes. Understanding the regulatory mechanisms of cardiomyocyte survival and death can identify potentially translatable therapeutic strategies for MI. Graphical abstract Highlights•Neonatal and juvenile mouse cardiomyocytes mainly undergo ferroptosis after MI•Cardiac fibroblasts protect cardiomyocytes through paracrine effect•Cardiac fibroblasts interact with cardiomyocytes to share iron burden•Pitx2 pathway protects cardiomyocytes from ferroptosis and controls fibrosis Cardiovascular medicine; Physiology; Cell biology
View Publication
Braam SR et al. (OCT 2009)
Trends in pharmacological sciences 30 10 536--45
Cardiomyocytes from human pluripotent stem cells in regenerative medicine and drug discovery.
Stem cells derived from pre-implantation human embryos or from somatic cells by reprogramming are pluripotent and self-renew indefinitely in culture. Pluripotent stem cells are unique in being able to differentiate to any cell type of the human body. Differentiation towards the cardiac lineage has attracted significant attention,initially with a strong focus on regenerative medicine. Although an important research area,the heart has proven challenging to repair by cardiomyocyte replacement. However,the ability to reprogramme adult cells to pluripotent stem cells and genetically manipulate stem cells presented opportunities to develop models of human disease. The availability of human cardiomyocytes from stem cell sources is expected to accelerate the discovery of cardiac drugs and safety pharmacology by offering more clinically relevant human culture models than presently available. Here we review the state-of-the-art using stem cell-derived human cardiomyocytes in drug discovery,drug safety pharmacology,and regenerative medicine.
View Publication
H. W. Grievink et al. ( 2022)
Frontiers in immunology 13 968815
Cardiovascular risk factors: The effects of ageing and smoking on the immune system, an observational clinical study.
Currently immunomodulatory compounds are under investigation for use in patients with cardiovascular disease,caused by atherosclerosis. These trials,using recurrent cardiovascular events as endpoint,require enrollment of large patient groups. We investigated the effect of key risk factors for atherosclerosis development,ageing and smoking,on the immune system,with the objective to identify biomarkers differentiating between human populations,and potentially serving as endpoints for future phase 1B trials with immunomodulatory compounds. Blood was collected from young healthy volunteers (aged 18-25 years,n=30),young smokers (18-25 years,n=20),elderly healthy volunteers (>60 years,n=20),heavy smokers (>45 years,15 packyears,n=11) and patients with stable coronary artery disease (CAD) (>60 years,n=27). Circulating immune cell subsets were characterized by flow cytometry,and collected plasma was evaluated by proteomics (Olink). Clear ageing effects were observed,mostly illustrated by a lower level in CD8+ and na{\{i}}ve CD4+ and CD8+ T cells with an increase in CD4+ and CD8+ effector memory T cells in elderly healthy volunteers compared to young healthy volunteers. Heavy smokers showed a more inflammatory cellular phenotype especially a shift in Th1/Th2 ratio: higher Th1 and lower Th2 percentages compared to young healthy volunteers. A significant decrease in circulating atheroprotective oxLDL-specific IgM was found in patients with CAD compared to young healthy volunteers. Elevated pro-inflammatory and chemotactic proteins TREM1 and CCL11 were observed in elderly volunteers compared to young volunteers. In addition heavy smokers had an increase in pro-inflammatory cytokine IL-6 and lysosomal protein LAMP3. These data show that ageing and smoking are associated with an inflammatory immunophenotype and that heavy smokers or aged individuals may serve as potential populations for future clinical trials investigating immunomodulatory drugs targeted for cardiovascular disease."
View Publication
W. Abplanalp et al. (jul 2019)
Journal of the American Heart Association 8 13 e013041
Carnosine Supplementation Mitigates the Deleterious Effects of Particulate Matter Exposure in Mice.
Background Exposure to fine airborne particulate matter ( PM 2.5) induces quantitative and qualitative defects in bone marrow-derived endothelial progenitor cells of mice,and similar outcomes in humans may contribute to vascular dysfunction and the cardiovascular morbidity and mortality associated with PM 2.5 exposure. Nevertheless,mechanisms underlying the pervasive effects of PM 2.5 are unclear and effective interventional strategies to mitigate against PM 2.5 toxicity are lacking. Furthermore,whether PM 2.5 exposure affects other types of bone marrow stem cells leading to additional hematological or immunological dysfunction is not clear. Methods and Results Mice given normal drinking water or that supplemented with carnosine,a naturally occurring,nucleophilic di-peptide that binds reactive aldehydes,were exposed to filtered air or concentrated ambient particles. Mice drinking normal water and exposed to concentrated ambient particles demonstrated a depletion of bone marrow hematopoietic stem cells but no change in mesenchymal stem cells. However,HSC depletion was significantly attenuated when the mice were placed on drinking water containing carnosine. Carnosine supplementation also increased the levels of carnosine-propanal conjugates in the urine of CAPs-exposed mice and prevented the concentrated ambient particles-induced dysfunction of endothelial progenitor cells as assessed by in vitro and in vivo assays. Conclusions These results suggest that exposure to PM 2.5 has pervasive effects on different bone marrow stem cell populations and that PM 2.5-induced hematopoietic stem cells depletion,endothelial progenitor cell dysfunction,and defects in vascular repair can be mitigated by excess carnosine. Carnosine supplementation may be a viable approach for preventing PM 2.5-induced immune dysfunction and cardiovascular injury in humans.
View Publication