Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 133(10)2023 05 15.
Article in English | MEDLINE | ID: mdl-37183820

ABSTRACT

Despite the prevalence of pericytes in the microvasculature of the heart, their role during ischemia-induced remodeling remains unclear. We used multiple lineage-tracing mouse models and found that pericytes migrated to the injury site and expressed profibrotic genes, coinciding with increased vessel leakage after myocardial infarction (MI). Single-cell RNA-Seq of cardiac pericytes at various time points after MI revealed the temporally regulated induction of genes related to vascular permeability, extracellular matrix production, basement membrane degradation, and TGF-ß signaling. Deleting TGF-ß receptor 1 in chondroitin sulfate proteoglycan 4-expressing (Cspg4-expressing) cells reduced fibrosis following MI, leading to a transient improvement in the cardiac ejection fraction. Furthermore, genetic ablation of Cspg4-expressing cells resulted in excessive vascular permeability, a decline in cardiac function, and increased mortality in the second week after MI. These data reveal an essential role for cardiac pericytes in the control of vascular homeostasis and the fibrotic response after acute ischemic injury, information that will help guide the development of novel strategies to preserve vascular integrity and attenuate pathological cardiac remodeling.


Subject(s)
Myocardial Infarction , Pericytes , Mice , Animals , Pericytes/metabolism , Myocardial Infarction/metabolism , Heart , Fibrosis , Extracellular Matrix/metabolism , Ventricular Remodeling/genetics , Myocardium/metabolism
2.
Nat Commun ; 12(1): 4155, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34230480

ABSTRACT

The organization of an integrated coronary vasculature requires the specification of immature endothelial cells (ECs) into arterial and venous fates based on their localization within the heart. It remains unclear how spatial information controls EC identity and behavior. Here we use single-cell RNA sequencing at key developmental timepoints to interrogate cellular contributions to coronary vessel patterning and maturation. We perform transcriptional profiling to define a heterogenous population of epicardium-derived cells (EPDCs) that express unique chemokine signatures. We identify a population of Slit2+ EPDCs that emerge following epithelial-to-mesenchymal transition (EMT), which we term vascular guidepost cells. We show that the expression of guidepost-derived chemokines such as Slit2 are induced in epicardial cells undergoing EMT, while mesothelium-derived chemokines are silenced. We demonstrate that epicardium-specific deletion of myocardin-related transcription factors in mouse embryos disrupts the expression of key guidance cues and alters EPDC-EC signaling, leading to the persistence of an immature angiogenic EC identity and inappropriate accumulation of ECs on the epicardial surface. Our study suggests that EC pathfinding and fate specification is controlled by a common mechanism and guided by paracrine signaling from EPDCs linking epicardial EMT to EC localization and fate specification in the developing heart.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/metabolism , Pericardium/cytology , Pericardium/metabolism , Animals , Chemokines , Coronary Vessels/metabolism , Embryo, Mammalian , Epithelial-Mesenchymal Transition , Gene Expression , Heart , Intercellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins , Nuclear Proteins , Pericardium/embryology , Serum Response Factor , Signal Transduction , Trans-Activators , Transcription Factors/metabolism , Transcriptome
4.
Cardiovasc Res ; 117(1): 201-211, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32176281

ABSTRACT

AIMS: Telomere attrition in cardiomyocytes is associated with decreased contractility, cellular senescence, and up-regulation of proapoptotic transcription factors. Pim1 is a cardioprotective kinase that antagonizes the aging phenotype of cardiomyocytes and delays cellular senescence by maintaining telomere length, but the mechanism remains unknown. Another pathway responsible for regulating telomere length is the transforming growth factor beta (TGFß) signalling pathway where inhibiting TGFß signalling maintains telomere length. The relationship between Pim1 and TGFß has not been explored. This study delineates the mechanism of telomere length regulation by the interplay between Pim1 and components of TGFß signalling pathways in proliferating A549 cells and post-mitotic cardiomyocytes. METHODS AND RESULTS: Telomere length was maintained by lentiviral-mediated overexpression of PIM1 and inhibition of TGFß signalling in A549 cells. Telomere length maintenance was further demonstrated in isolated cardiomyocytes from mice with cardiac-specific overexpression of PIM1 and by pharmacological inhibition of TGFß signalling. Mechanistically, Pim1 inhibited phosphorylation of Smad2, preventing its translocation into the nucleus and repressing expression of TGFß pathway genes. CONCLUSION: Pim1 maintains telomere lengths in cardiomyocytes by inhibiting phosphorylation of the TGFß pathway downstream effectors Smad2 and Smad3, which prevents repression of telomerase reverse transcriptase. Findings from this study demonstrate a novel mechanism of telomere length maintenance and provide a potential target for preserving cardiac function.


Subject(s)
Cellular Senescence/drug effects , Myocytes, Cardiac/drug effects , Proto-Oncogene Proteins c-pim-1/metabolism , Telomere Homeostasis/drug effects , Transforming Growth Factor beta1/pharmacology , A549 Cells , Animals , Humans , Male , Mice, Knockout , Myocytes, Cardiac/enzymology , Phosphorylation , Proto-Oncogene Proteins c-pim-1/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Telomerase/metabolism
5.
Circ Res ; 126(3): 377-394, 2020 01 31.
Article in English | MEDLINE | ID: mdl-31999538

ABSTRACT

The heart is lined by a single layer of mesothelial cells called the epicardium that provides important cellular contributions for embryonic heart formation. The epicardium harbors a population of progenitor cells that undergo epithelial-to-mesenchymal transition displaying characteristic conversion of planar epithelial cells into multipolar and invasive mesenchymal cells before differentiating into nonmyocyte cardiac lineages, such as vascular smooth muscle cells, pericytes, and fibroblasts. The epicardium is also a source of paracrine cues that are essential for fetal cardiac growth, coronary vessel patterning, and regenerative heart repair. Although the epicardium becomes dormant after birth, cardiac injury reactivates developmental gene programs that stimulate epithelial-to-mesenchymal transition; however, it is not clear how the epicardium contributes to disease progression or repair in the adult. In this review, we will summarize the molecular mechanisms that control epicardium-derived progenitor cell migration, and the functional contributions of the epicardium to heart formation and cardiomyopathy. Future perspectives will be presented to highlight emerging therapeutic strategies aimed at harnessing the regenerative potential of the fetal epicardium for cardiac repair.


Subject(s)
Heart Diseases/etiology , Pericardium/growth & development , Regeneration , Animals , Humans , Myocardium/cytology , Myocardium/metabolism , Paracrine Communication , Pericardium/cytology , Pericardium/metabolism , Pericardium/physiology
6.
Commun Biol ; 2: 205, 2019.
Article in English | MEDLINE | ID: mdl-31231694

ABSTRACT

Cardiomyocyte ploidy has been described but remains obscure in cardiac interstitial cells. Ploidy of c-kit+ cardiac interstitial cells was assessed using confocal, karyotypic, and flow cytometric technique. Notable differences were found between rodent (rat, mouse) c-kit+ cardiac interstitial cells possessing mononuclear tetraploid (4n) content, compared to large mammals (human, swine) with mononuclear diploid (2n) content. In-situ analysis, confirmed with fresh isolates, revealed diploid content in human c-kit+ cardiac interstitial cells and a mixture of diploid and tetraploid content in mouse. Downregulation of the p53 signaling pathway provides evidence why rodent, but not human, c-kit+ cardiac interstitial cells escape replicative senescence. Single cell transcriptional profiling reveals distinctions between diploid versus tetraploid populations in mouse c-kit+ cardiac interstitial cells, alluding to functional divergences. Collectively, these data reveal notable species-specific biological differences in c-kit+ cardiac interstitial cells, which could account for challenges in extrapolation of myocardial from preclinical studies to clinical trials.


Subject(s)
Cellular Senescence , Myocytes, Cardiac/metabolism , Signal Transduction , Stem Cells/cytology , Tetraploidy , Animals , Cell Proliferation , Down-Regulation , Female , Flow Cytometry , Gene Expression Profiling , Humans , Karyotyping , Leukocytes, Mononuclear/cytology , Male , Mice , Microscopy, Confocal , Ploidies , Rats , Swine
7.
JCI Insight ; 4(5)2019 03 07.
Article in English | MEDLINE | ID: mdl-30702442

ABSTRACT

ß-2 Microglobulin (ß2M) is a molecular chaperone for the major histocompatibility class I (MHC I) complex, hemochromatosis factor protein (HFE), and the neonatal Fc receptor (FcRn), but ß2M may also have less understood chaperone-independent functions. Elevated plasma ß2M has a direct role in neurocognitive decline and is a risk factor for adverse cardiovascular events. ß2M mRNA is present in platelets at very high levels, and ß2M is part of the activated platelet releasate. In addition to their more well-studied thrombotic functions, platelets are important immune regulatory cells that release inflammatory molecules and contribute to leukocyte trafficking, activation, and differentiation. We have now found that platelet-derived ß2M is a mediator of monocyte proinflammatory differentiation through noncanonical TGFß receptor signaling. Circulating monocytes from mice lacking ß2M only in platelets (Plt-ß2M-/-) had a more proreparative monocyte phenotype, in part dependent on increased platelet-derived TGFß signaling in the absence of ß2M. Using a mouse myocardial infarction (MI) model, Plt-ß2M-/- mice had limited post-MI proinflammatory monocyte responses and, instead, demonstrated early proreparative monocyte differentiation, profibrotic myofibroblast responses, and a rapid decline in heart function compared with WT mice. These data demonstrate a potentially novel chaperone-independent, monocyte phenotype-regulatory function for platelet ß2M and that platelet-derived 2M and TGFß have opposing roles in monocyte differentiation that may be important in tissue injury responses.


Subject(s)
Blood Platelets/metabolism , Monocytes/metabolism , beta 2-Microglobulin/metabolism , Animals , Cell Differentiation , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Chaperones , Platelet Activation , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , THP-1 Cells , beta 2-Microglobulin/genetics
8.
J Mol Cell Cardiol ; 129: 92-104, 2019 04.
Article in English | MEDLINE | ID: mdl-30771308

ABSTRACT

Serum response factor (SRF) and the SRF co-activators myocardin-related transcription factors (MRTFs) are essential for epicardium-derived progenitor cell (EPDC)-mobilization during heart development; however, the impact of developmental EPDC deficiencies on adult cardiac physiology has not been evaluated. Here, we utilize the Wilms Tumor-1 (Wt1)-Cre to delete Mrtfs or Srf in the epicardium, which reduced the number of EPDCs in the adult cardiac interstitium. Deficiencies in Wt1-lineage EPDCs prevented the development of cardiac fibrosis and diastolic dysfunction in aged mice. Mice lacking MRTF or SRF in EPDCs also displayed preservation of cardiac function following myocardial infarction partially due to the depletion of Wt1 lineage-derived cells in the infarct. Interestingly, depletion of Wt1-lineage EPDCs allows for the population of the infarct with a Wt1-negative cell lineage with a reduced fibrotic profile. Taken together, our study conclusively demonstrates the contribution of EPDCs to both ischemic cardiac remodeling and the development of diastolic dysfunction in old age, and reveals the existence of an alternative Wt1-negative source of resident fibroblasts that can populate the infarct.


Subject(s)
Aging/pathology , Fibroblasts/pathology , Myocardial Ischemia/pathology , Pericardium/pathology , Animals , Cell Lineage , Diastole , Fibrosis , Heart/physiopathology , Mice, Knockout , Myocardial Ischemia/physiopathology , Serum Response Factor/metabolism , Stem Cells/metabolism , Trans-Activators/metabolism , Ventricular Remodeling , WT1 Proteins/metabolism
9.
J Mol Cell Cardiol ; 127: 154-164, 2019 02.
Article in English | MEDLINE | ID: mdl-30571978

ABSTRACT

RATIONALE: Understanding and manipulating the cardiomyocyte cell cycle has been the focus of decades of research, however the ultimate goal of activating mitotic activity in adult mammalian cardiomyocytes remains elusive and controversial. The relentless pursuit of controlling cardiomyocyte mitosis has been complicated and obfuscated by a multitude of indices used as evidence of cardiomyocyte cell cycle activity that lack clear identification of cardiomyocyte "proliferation" versus cell cycle progression, endoreplication, endomitosis, and even DNA damage. Unambiguous appreciation of the complexity of cardiomyocyte replication that avoids oversimplification and misinterpretation is desperately needed. OBJECTIVE: Track cardiomyocyte cell cycle activity and authenticate fidelity of proliferation markers as indicators of de novo cardiomyogenesis in post-mitotic cardiomyocytes. METHODS AND RESULTS: Cardiomyocytes expressing the FUCCI construct driven by the α-myosin heavy chain promoter were readily and uniformly detected through the myocardium of transgenic mice. Cardiomyocyte cell cycle activity peaks at postnatal day 2 and rapidly declines thereafter with almost all cardiomyocytes arrested at the G1/S cell cycle transition. Myocardial infarction injury in adult hearts prompts transient small increases in myocytes progressing through cell cycle without concurrent mitotic activity, indicating lack of cardiomyogenesis. In comparison, cardiomyogenic activity during early postnatal development correlated with coincidence of FUCCI and cKit+ cells that were undetectable in the adult myocardium. CONCLUSIONS: Cardiomyocyte-specific expression of Fluorescence Ubiquitination-based Cell Cycle Indicators (FUCCI) reveals previously unappreciated aspects of cardiomyocyte cell cycle arrest and biological activity in postnatal development and in response to pathologic damage. Compared to many other methods and model systems, the FUCCI transgenic (FUCCI-Tg) mouse represents a valuable tool to unambiguously track cell cycle and proliferation of the entire cardiomyocyte population in the adult murine heart. FUCCI-Tg provides a desperately needed novel approach in the armamentarium of tools to validate cardiomyocyte proliferative activity that will reveal cell cycle progression, discriminate between cycle progression, DNA replication, and proliferation, and provide important insight for enhancing cardiomyocyte proliferation in the context of adult myocardial tissue.


Subject(s)
Cell Cycle , Gene Transfer Techniques , Heart/physiology , Myocytes, Cardiac/cytology , Ubiquitination , Animals , Animals, Newborn , Cell Cycle Checkpoints , Cell Division , Cell Proliferation , Cells, Cultured , Fluorescence , Mice, Transgenic , Organ Specificity
10.
Circulation ; 138(17): 1864-1878, 2018 10 23.
Article in English | MEDLINE | ID: mdl-29716942

ABSTRACT

BACKGROUND: Hypertrophic cardiomyocyte growth and dysfunction accompany various forms of heart disease. The mechanisms responsible for transcriptional changes that affect cardiac physiology and the transition to heart failure are not well understood. The intercalated disc (ID) is a specialized intercellular junction coupling cardiomyocyte force transmission and propagation of electrical activity. The ID is gaining attention as a mechanosensitive signaling hub and hotspot for causative mutations in cardiomyopathy. METHODS: Transmission electron microscopy, confocal microscopy, and single-molecule localization microscopy were used to examine changes in ID structure and protein localization in the murine and human heart. We conducted detailed cardiac functional assessment and transcriptional profiling of mice lacking myocardin-related transcription factor (MRTF)-A and MRTF-B specifically in adult cardiomyocytes to evaluate the role of mechanosensitive regulation of gene expression in load-induced ventricular remodeling. RESULTS: We found that MRTFs localize to IDs in the healthy human heart and accumulate in the nucleus in heart failure. Although mice lacking MRTFs in adult cardiomyocytes display normal cardiac physiology at baseline, pressure overload leads to rapid heart failure characterized by sarcomere disarray, ID disintegration, chamber dilation and wall thinning, cardiac functional decline, and partially penetrant acute lethality. Transcriptional profiling reveals a program of actin cytoskeleton and cardiomyocyte adhesion genes driven by MRTFs during pressure overload. Indeed, conspicuous remodeling of gap junctions at IDs identified by single-molecule localization microscopy may partially stem from a reduction in Mapre1 expression, which we show is a direct mechanosensitive MRTF target. CONCLUSIONS: Our study describes a novel paradigm in which MRTFs control an acute mechanosensitive signaling circuit that coordinates cross-talk between the actin and microtubule cytoskeleton and maintains ID integrity and cardiomyocyte homeostasis in heart disease.


Subject(s)
Heart Failure/metabolism , Hypertrophy, Left Ventricular/metabolism , Mechanotransduction, Cellular , Myocytes, Cardiac/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Aged , Animals , Animals, Newborn , COS Cells , Case-Control Studies , Chlorocebus aethiops , Connexin 43/genetics , Connexin 43/metabolism , Female , Gene Expression Regulation , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Middle Aged , Myocytes, Cardiac/ultrastructure , NIH 3T3 Cells , Single Molecule Imaging , Trans-Activators/deficiency , Trans-Activators/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , Ventricular Function, Left , Ventricular Remodeling
11.
Proc Natl Acad Sci U S A ; 115(15): E3436-E3445, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29581288

ABSTRACT

Heart disease is associated with the accumulation of resident cardiac fibroblasts (CFs) that secrete extracellular matrix (ECM), leading to the development of pathological fibrosis and heart failure. However, the mechanisms underlying resident CF proliferation remain poorly defined. Here, we report that small proline-rich protein 2b (Sprr2b) is among the most up-regulated genes in CFs during heart disease. We demonstrate that SPRR2B is a regulatory subunit of the USP7/MDM2-containing ubiquitination complex. SPRR2B stimulates the accumulation of MDM2 and the degradation of p53, thus facilitating the proliferation of pathological CFs. Furthermore, SPRR2B phosphorylation by nonreceptor tyrosine kinases in response to TGF-ß1 signaling and free-radical production potentiates SPRR2B activity and cell cycle progression. Knockdown of the Sprr2b gene or inhibition of SPRR2B phosphorylation attenuates USP7/MDM2 binding and p53 degradation, leading to CF cell cycle arrest. Importantly, SPRR2B expression is elevated in cardiac tissue from human heart failure patients and correlates with the proliferative state of patient-derived CFs in a process that is reversed by insulin growth factor-1 signaling. These data establish SPRR2B as a unique component of the USP7/MDM2 ubiquitination complex that drives p53 degradation, CF accumulation, and the development of pathological cardiac fibrosis.


Subject(s)
Cell Proliferation , Cornified Envelope Proline-Rich Proteins/metabolism , Fibroblasts/metabolism , Heart Failure/metabolism , Tumor Suppressor Protein p53/metabolism , Adult , Aged , Animals , Cornified Envelope Proline-Rich Proteins/genetics , Heart Failure/genetics , Heart Failure/physiopathology , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Myocardium/metabolism , Proteolysis , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Tumor Suppressor Protein p53/genetics
12.
Stem Cells ; 36(6): 868-880, 2018 06.
Article in English | MEDLINE | ID: mdl-29441645

ABSTRACT

Aging severely limits myocardial repair and regeneration. Delineating the impact of age-associated factors such as short telomeres is critical to enhance the regenerative potential of cardiac progenitor cells (CPCs). We hypothesized that short telomeres activate p53 and induce autophagy to elicit the age-associated change in CPC fate. We isolated CPCs and compared mouse strains with different telomere lengths for phenotypic characteristics of aging. Wild mouse strain Mus musculus castaneus (CAST) possessing short telomeres exhibits early cardiac aging with cardiac dysfunction, hypertrophy, fibrosis, and senescence, as compared with common lab strains FVB and C57 bearing longer telomeres. CAST CPCs with short telomeres demonstrate altered cell fate as characterized by cell cycle arrest, senescence, basal commitment, and loss of quiescence. Elongation of telomeres using a modified mRNA for telomerase restores youthful properties to CAST CPCs. Short telomeres induce autophagy in CPCs, a catabolic protein degradation process, as evidenced by reduced p62 and increased accumulation of autophagic puncta. Pharmacological inhibition of autophagosome formation reverses the cell fate to a more youthful phenotype. Mechanistically, cell fate changes induced by short telomeres are partially p53 dependent, as p53 inhibition rescues senescence and commitment observed in CAST CPCs, coincident with attenuation of autophagy. In conclusion, short telomeres activate p53 and autophagy to tip the equilibrium away from quiescence and proliferation toward differentiation and senescence, leading to exhaustion of CPCs. This study provides the mechanistic basis underlying age-associated cell fate changes that will enable identification of molecular strategies to prevent senescence of CPCs. Stem Cells 2018;36:868-880.


Subject(s)
Heart/physiology , Stem Cells/metabolism , Telomere Shortening/physiology , Telomere/metabolism , Tumor Suppressor Protein p53/metabolism , Aging , Animals , Autophagy , Cell Differentiation , Humans , Mice
13.
PLoS One ; 12(3): e0173963, 2017.
Article in English | MEDLINE | ID: mdl-28323876

ABSTRACT

PIM1, a pro-survival gene encoding a serine/ threonine kinase, influences cell proliferation and survival. Modification of cardiac progenitor cells (CPCs) or cardiomyocytes with PIM1 using a lentivirus-based delivery method showed long-term improved cardiac function after myocardial infarction (MI). However, lentivirus based delivery methods have stringent FDA regulation with respect to clinical trials. To provide an alternative and low risk PIM1 delivery method, this study examined the use of a non-viral modified plasmid-minicircle (MC) as a vehicle to deliver PIM1 into mouse CPCs (mCPCs) in vitro and the myocardium in vivo. MC containing a turbo gfp reporter gene (gfp-MC) was used as a transfection and injection control. PIM1 was subcloned into gfp-MC (PIM1-MC) and then transfected into mCPCs at an efficiency of 29.4±3.7%. PIM1-MC engineered mCPCs (PIM1-mCPCs) exhibit significantly (P<0.05) better survival rate under oxidative treatment. PIM1-mCPCs also exhibit 1.9±0.1 and 2.2±0.2 fold higher cell proliferation at 3 and 5 days post plating, respectively, as compared to gfp-MC transfected mCPCs control. PIM1-MC was injected directly into ten-week old adult FVB female mice hearts in the border zone immediately after MI. Delivery of PIM1 into myocardium was confirmed by GFP+ cardiomyocytes. Mice with PIM1-MC injection showed increased protection compared to gfp-MC injection groups measured by ejection fraction at 3 and 7 days post injury (P = 0.0379 and P = 0.0262 by t-test, respectively). Success of PIM1 delivery and integration into mCPCs in vitro and cardiomyocytes in vivo by MC highlights the possibility of a non-cell based therapeutic approach for treatment of ischemic heart disease and MI.


Subject(s)
Genetic Therapy/methods , Myocardial Infarction/genetics , Myocardial Infarction/therapy , Proto-Oncogene Proteins c-pim-1/genetics , Animals , Cell Proliferation , Cell Survival , Cells, Cultured , Disease Models, Animal , Female , Genetic Vectors , Mice , Myoblasts, Cardiac/metabolism , Myoblasts, Cardiac/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Plasmids/genetics , Transfection
14.
J Mol Cell Cardiol ; 100: 54-63, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27721024

ABSTRACT

BACKGROUND: Myocardial infarction is followed by cardiac dysfunction, cellular death, and ventricular remodeling, including tissue fibrosis. S100A4 protein plays multiple roles in cellular survival, and tissue fibrosis, but the relative role of the S100A4 in the myocardium after myocardial infarction is unknown. This study aims to investigate the role of S100A4 in myocardial remodeling and cardiac function following infarct damage. METHODS AND RESULTS: S100A4 expression is low in the adult myocardium, but significantly increased following myocardial infarction. Deletion of S100A4 increased cardiac damage after myocardial infarction, whereas cardiac myocyte-specific overexpression of S100A4 protected the infarcted myocardium. Decreased cardiac function in S100A4 Knockout mice was accompanied with increased cardiac remodeling, fibrosis, and diminished capillary density in the remote myocardium. Loss of S100A4 caused increased apoptotic cell death both in vitro and in vivo in part mediated by decreased VEGF expression. Conversely, S100A4 overexpression protected cells against apoptosis in vitro and in vivo. Increased pro-survival AKT-signaling explained reduced apoptosis in S100A4 overexpressing cells. CONCLUSION: S100A4 expression protects cardiac myocytes against myocardial ischemia and is required for stabilization of cardiac function after MI.


Subject(s)
Myocardial Ischemia/genetics , Myocardial Ischemia/metabolism , Myocardium/metabolism , S100 Calcium-Binding Protein A4/genetics , Stress, Physiological/genetics , Animals , Cell Death/genetics , Disease Models, Animal , Echocardiography , Gene Expression , Hemodynamics , Mice , Mice, Knockout , Myocardial Infarction/diagnosis , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardial Ischemia/diagnosis , Myocardial Ischemia/physiopathology , Myocardium/pathology , S100 Calcium-Binding Protein A4/metabolism , Ventricular Remodeling
15.
J Biol Chem ; 290(42): 25411-26, 2015 Oct 16.
Article in English | MEDLINE | ID: mdl-26324717

ABSTRACT

Ca(2+)/Calmodulin-dependent protein kinase II (CaMKII) signaling in the heart regulates cardiomyocyte contractility and growth in response to elevated intracellular Ca(2+). The δB isoform of CaMKII is the predominant nuclear splice variant in the adult heart and regulates cardiomyocyte hypertrophic gene expression by signaling to the histone deacetylase HDAC4. However, the role of CaMKIIδ in cardiac progenitor cells (CPCs) has not been previously explored. During post-natal growth endogenous CPCs display primarily cytosolic CaMKIIδ, which localizes to the nuclear compartment of CPCs after myocardial infarction injury. CPCs undergoing early differentiation in vitro increase levels of CaMKIIδB in the nuclear compartment where the kinase may contribute to the regulation of CPC commitment. CPCs modified with lentiviral-based constructs to overexpress CaMKIIδB (CPCeδB) have reduced proliferative rate compared with CPCs expressing eGFP alone (CPCe). Additionally, stable expression of CaMKIIδB promotes distinct morphological changes such as increased cell surface area and length of cells compared with CPCe. CPCeδB are resistant to oxidative stress induced by hydrogen peroxide (H2O2) relative to CPCe, whereas knockdown of CaMKIIδB resulted in an up-regulation of cell death and cellular senescence markers compared with scrambled treated controls. Dexamethasone (Dex) treatment increased mRNA and protein expression of cardiomyogenic markers cardiac troponin T and α-smooth muscle actin in CPCeδB compared with CPCe, suggesting increased differentiation. Therefore, CaMKIIδB may serve as a novel modulatory protein to enhance CPC survival and commitment into the cardiac and smooth muscle lineages.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Lineage , Cell Nucleus/enzymology , Cell Survival , Isoenzymes/metabolism , Myocytes, Cardiac/cytology , Signal Transduction , Stem Cells/cytology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Gene Knockdown Techniques , Isoenzymes/genetics , Male , Mice , Myocytes, Cardiac/enzymology , Stem Cells/enzymology
16.
Circ Res ; 117(8): 695-706, 2015 Sep 25.
Article in English | MEDLINE | ID: mdl-26228030

ABSTRACT

RATIONALE: Dual cell transplantation of cardiac progenitor cells (CPCs) and mesenchymal stem cells (MSCs) after infarction improves myocardial repair and performance in large animal models relative to delivery of either cell population. OBJECTIVE: To demonstrate that CardioChimeras (CCs) formed by fusion between CPCs and MSCs have enhanced reparative potential in a mouse model of myocardial infarction relative to individual stem cells or combined cell delivery. METHODS AND RESULTS: Two distinct and clonally derived CCs, CC1 and CC2, were used for this study. CCs improved left ventricular anterior wall thickness at 4 weeks post injury, but only CC1 treatment preserved anterior wall thickness at 18 weeks. Ejection fraction was enhanced at 6 weeks in CCs, and functional improvements were maintained in CCs and CPC+MSC groups at 18 weeks. Infarct size was decreased in CCs, whereas CPC+MSC and CPC parent groups remained unchanged at 12 weeks. CCs exhibited increased persistence, engraftment, and expression of early commitment markers within the border zone relative to combinatorial and individual cell population-injected groups. CCs increased capillary density and preserved cardiomyocyte size in the infarcted regions suggesting CCs role in protective paracrine secretion. CONCLUSIONS: CCs merge the application of distinct cells into a single entity for cellular therapeutic intervention in the progression of heart failure. CCs are a novel cell therapy that improves on combinatorial cell approaches to support myocardial regeneration.


Subject(s)
Anterior Wall Myocardial Infarction/surgery , Heart Ventricles/physiopathology , Mesenchymal Stem Cell Transplantation , Myocytes, Cardiac/transplantation , Regeneration , Transplantation Chimera , Animals , Animals, Newborn , Anterior Wall Myocardial Infarction/metabolism , Anterior Wall Myocardial Infarction/pathology , Anterior Wall Myocardial Infarction/physiopathology , Biomarkers/metabolism , Cell Proliferation , Cell Size , Cell Survival , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Female , Graft Survival , Heart Ventricles/metabolism , Heart Ventricles/pathology , Mice , Myocardial Contraction , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Neovascularization, Physiologic , Paracrine Communication , Phenotype , Rats , Recovery of Function , Stroke Volume , Time Factors , Transfection , Ventricular Function, Left
17.
J Pathol ; 237(4): 482-94, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26213100

ABSTRACT

The low molecular weight protein tyrosine phosphatase (LMPTP), encoded by the ACP1 gene, is a ubiquitously expressed phosphatase whose in vivo function in the heart and in cardiac diseases remains unknown. To investigate the in vivo role of LMPTP in cardiac function, we generated mice with genetic inactivation of the Acp1 locus and studied their response to long-term pressure overload. Acp1(-/-) mice develop normally and ageing mice do not show pathology in major tissues under basal conditions. However, Acp1(-/-) mice are strikingly resistant to pressure overload hypertrophy and heart failure. Lmptp expression is high in the embryonic mouse heart, decreased in the postnatal stage, and increased in the adult mouse failing heart. We also show that LMPTP expression increases in end-stage heart failure in humans. Consistent with their protected phenotype, Acp1(-/-) mice subjected to pressure overload hypertrophy have attenuated fibrosis and decreased expression of fibrotic genes. Transcriptional profiling and analysis of molecular signalling show that the resistance of Acp1(-/-) mice to pathological cardiac stress correlates with marginal re-expression of fetal cardiac genes, increased insulin receptor beta phosphorylation, as well as PKA and ephrin receptor expression, and inactivation of the CaMKIIδ pathway. Our data show that ablation of Lmptp inhibits pathological cardiac remodelling and suggest that inhibition of LMPTP may be of therapeutic relevance for the treatment of human heart failure.


Subject(s)
Heart Failure/metabolism , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins/metabolism , Takotsubo Cardiomyopathy/metabolism , Animals , Disease Models, Animal , Fluorescent Antibody Technique , Humans , Immunoblotting , Immunoprecipitation , Mice , Mice, Inbred BALB C , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Rats
18.
Basic Res Cardiol ; 110(3): 29, 2015 May.
Article in English | MEDLINE | ID: mdl-25893875

ABSTRACT

Phase I clinical trials applying autologous progenitor cells to treat heart failure have yielded promising results; however, improvement in function is modest, indicating a need to enhance cardiac stem cell reparative capacity. Notch signaling plays a crucial role in cardiac development, guiding cell fate decisions that underlie myocyte and vessel differentiation. The Notch pathway is retained in the adult cardiac stem cell niche, where level and duration of Notch signal influence proliferation and differentiation of cardiac progenitors. In this study, Notch signaling promotes growth, survival and differentiation of cardiac progenitor cells into smooth muscle lineages in vitro. Cardiac progenitor cells expressing tamoxifen-regulated intracellular Notch1 (CPCeK) are significantly larger and proliferate more slowly than control cells, exhibit elevated mTORC1 and Akt signaling, and are resistant to oxidative stress. Vascular smooth muscle and cardiomyocyte markers increase in CPCeK and are augmented further upon ligand-mediated induction of Notch signal. Paracrine signals indicative of growth, survival and differentiation increase with Notch activity, while markers of senescence are decreased. Adoptive transfer of CPCeK into infarcted mouse myocardium enhances preservation of cardiac function and reduces infarct size relative to hearts receiving control cells. Greater capillary density and proportion of vascular smooth muscle tissue in CPCeK-treated hearts indicate improved vascularization. Finally, we report a previously undescribed signaling mechanism whereby Notch activation stimulates CPC growth, survival and differentiation via mTORC1 and paracrine factor expression. Taken together, these findings suggest that regulated Notch activation potentiates the reparative capacity of CPCs in the treatment of cardiac disease.


Subject(s)
Cell Differentiation/physiology , Cell- and Tissue-Based Therapy/methods , Myocardial Infarction/therapy , Myocytes, Cardiac/cytology , Receptors, Notch/metabolism , Stem Cell Transplantation/methods , Adoptive Transfer , Animals , Cell Lineage , Disease Models, Animal , Immunoblotting , Immunohistochemistry , Mice , Myocytes, Cardiac/metabolism , Real-Time Polymerase Chain Reaction , Stem Cells/cytology , Stem Cells/metabolism
19.
J Biol Chem ; 290(22): 13935-47, 2015 May 29.
Article in English | MEDLINE | ID: mdl-25882843

ABSTRACT

Human cardiac progenitor cells (hCPC) improve heart function after autologous transfer in heart failure patients. Regenerative potential of hCPCs is severely limited with age, requiring genetic modification to enhance therapeutic potential. A legacy of work from our laboratory with Pim1 kinase reveals effects on proliferation, survival, metabolism, and rejuvenation of hCPCs in vitro and in vivo. We demonstrate that subcellular targeting of Pim1 bolsters the distinct cardioprotective effects of this kinase in hCPCs to increase proliferation and survival, and antagonize cellular senescence. Adult hCPCs isolated from patients undergoing left ventricular assist device implantation were engineered to overexpress Pim1 throughout the cell (PimWT) or targeted to either mitochondrial (Mito-Pim1) or nuclear (Nuc-Pim1) compartments. Nuc-Pim1 enhances stem cell youthfulness associated with decreased senescence-associated ß-galactosidase activity, preserved telomere length, reduced expression of p16 and p53, and up-regulation of nucleostemin relative to PimWT hCPCs. Alternately, Mito-Pim1 enhances survival by increasing expression of Bcl-2 and Bcl-XL and decreasing cell death after H2O2 treatment, thereby preserving mitochondrial integrity superior to PimWT. Mito-Pim1 increases the proliferation rate by up-regulation of cell cycle modulators Cyclin D, CDK4, and phospho-Rb. Optimal stem cell traits such as proliferation, survival, and increased youthful properties of aged hCPCs are enhanced after targeted Pim1 localization to mitochondrial or nuclear compartments. Targeted Pim1 overexpression in hCPCs allows for selection of the desired phenotypic properties to overcome patient variability and improve specific stem cell characteristics.


Subject(s)
Gene Expression Regulation , Heart/physiology , Proto-Oncogene Proteins c-pim-1/metabolism , Stem Cells/metabolism , Apoptosis , Cell Cycle , Cell Nucleus/metabolism , Cell Proliferation , Cell Survival , Cellular Senescence , Green Fluorescent Proteins/metabolism , Heart Failure , Heart Ventricles/metabolism , Humans , Lentivirus/metabolism , Mitochondria/metabolism , Myocardium/cytology , Myocardium/metabolism , Phenotype , Regeneration , Stem Cells/cytology , Subcellular Fractions/metabolism , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...