Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Am J Physiol Heart Circ Physiol ; 314(1): H68-H81, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28939651

ABSTRACT

Notch receptor signaling is active during cardiac development and silenced in myocytes after birth. Conversely, outward K+ Kv currents progressively appear in postnatal myocytes leading to shortening of the action potential (AP) and acquisition of the mature electrical phenotype. In the present study, we tested the possibility that Notch signaling modulates the electrical behavior of cardiomyocytes by interfering with Kv currents. For this purpose, the effects of Notch receptor activity on electrophysiological properties of myocytes were evaluated using transgenic mice with inducible expression of the Notch1 intracellular domain (NICD), the functional fragment of the activated Notch receptor, and in neonatal myocytes after inhibition of the Notch transduction pathway. By patch clamp, NICD-overexpressing cells presented prolonged AP duration and reduced upstroke amplitude, properties that were coupled with reduced rapidly activating Kv and fast Na+ currents, compared with cells obtained from wild-type mice. In cultured neonatal myocytes, inhibition of the proteolitic release of NICD with a γ-secretase antagonist increased transcript levels of the Kv channel-interacting proteins 2 (KChIP2) and enhanced the density of Kv currents. Collectively, these results indicate that Notch signaling represents an important regulator of the electrophysiological behavior of developing and adult myocytes by repressing, at least in part, repolarizing Kv currents. NEW & NOTEWORTHY We investigated the effects of Notch receptor signaling on the electrical properties of cardiomyocytes. Our results indicate that the Notch transduction pathway interferes with outward K+ Kv currents, critical determinants of the electrical repolarization of myocytes.


Subject(s)
Myocytes, Cardiac/metabolism , Potassium Channels, Voltage-Gated/metabolism , Potassium/metabolism , Receptor, Notch1/metabolism , Signal Transduction , Animals , Cells, Cultured , Female , Kinetics , Kv Channel-Interacting Proteins/genetics , Kv Channel-Interacting Proteins/metabolism , Male , Membrane Potentials , Mice, Inbred C57BL , Mice, Transgenic , Potassium Channels, Voltage-Gated/genetics , Receptor, Notch1/genetics , Sodium/metabolism
2.
Am J Physiol Heart Circ Physiol ; 312(1): H150-H161, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27881388

ABSTRACT

Diabetes and other metabolic conditions characterized by elevated blood glucose constitute important risk factors for cardiovascular disease. Hyperglycemia targets myocardial cells rendering ineffective mechanical properties of the heart, but cellular alterations dictating the progressive deterioration of cardiac function with metabolic disorders remain to be clarified. In the current study, we examined the effects of hyperglycemia on cardiac function and myocyte physiology by employing mice with high blood glucose induced by administration of streptozotocin, a compound toxic to insulin-producing ß-cells. We found that hyperglycemia initially delayed the electrical recovery of the heart, whereas cardiac function became defective only after ~2 mo with this condition and gradually worsened with time. Prolonged hyperglycemia was associated with increased chamber dilation, thinning of the left ventricle (LV), and myocyte loss. Cardiomyocytes from hyperglycemic mice exhibited defective Ca2+ transients before the appearance of LV systolic defects. Alterations in Ca2+ transients involved enhanced spontaneous Ca2+ releases from the sarcoplasmic reticulum (SR), reduced cytoplasmic Ca2+ clearance, and declined SR Ca2+ load. These defects have important consequences on myocyte contraction, relaxation, and mechanisms of rate adaptation. Collectively, our data indicate that hyperglycemia alters intracellular Ca2+ homeostasis in cardiomyocytes, hindering contractile activity and contributing to the manifestation of the diabetic cardiomyopathy. NEW & NOTEWORTHY: We have investigated the effects of hyperglycemia on cardiomyocyte physiology and ventricular function. Our results indicate that defective Ca2+ handling is a critical component of the progressive deterioration of cardiac performance of the diabetic heart.


Subject(s)
Calcium/metabolism , Diabetes Mellitus, Experimental/metabolism , Homeostasis , Hyperglycemia/metabolism , Myocytes, Cardiac/metabolism , Ventricular Dysfunction, Left/physiopathology , Action Potentials , Animals , Caffeine/pharmacology , Central Nervous System Stimulants/pharmacology , Diabetes Mellitus, Experimental/complications , Echocardiography , Electrocardiography , Female , Isolated Heart Preparation , Male , Mice , Myocytes, Cardiac/drug effects , Patch-Clamp Techniques , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/etiology
3.
Circ Res ; 116(1): 150-66, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25552694

ABSTRACT

This review article discusses the mechanisms of cardiomyogenesis in the adult heart. They include the re-entry of cardiomyocytes into the cell cycle; dedifferentiation of pre-existing cardiomyocytes, which assume an immature replicating cell phenotype; transdifferentiation of hematopoietic stem cells into cardiomyocytes; and cardiomyocytes derived from activation and lineage specification of resident cardiac stem cells. The recognition of the origin of cardiomyocytes is of critical importance for the development of strategies capable of enhancing the growth response of the myocardium; in fact, cell therapy for the decompensated heart has to be based on the acquisition of this fundamental biological knowledge.


Subject(s)
Heart/growth & development , Hematopoietic Stem Cells/physiology , Myocytes, Cardiac/physiology , Organogenesis/physiology , Adult , Animals , Cell Differentiation/physiology , Humans
4.
Am J Physiol Heart Circ Physiol ; 310(7): H873-90, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26801307

ABSTRACT

Studies of myocardial aging are complex and the mechanisms involved in the deterioration of ventricular performance and decreased functional reserve of the old heart remain to be properly defined. We have studied a colony of beagle dogs from 3 to 14 yr of age kept under a highly regulated environment to define the effects of aging on the myocardium. Ventricular, myocardial, and myocyte function, together with anatomical and structural properties of the organ and cardiomyocytes, were evaluated. Ventricular hypertrophy was not observed with aging and the structural composition of the myocardium was modestly affected. Alterations in the myocyte compartment were identified in aged dogs, and these factors negatively interfere with the contractile reserve typical of the young heart. The duration of the action potential is prolonged in old cardiomyocytes contributing to the slower electrical recovery of the myocardium. Also, the remodeled repolarization of cardiomyocytes with aging provides inotropic support to the senescent muscle but compromises its contractile reserve, rendering the old heart ineffective under conditions of high hemodynamic demand. The defects in the electrical and mechanical properties of cardiomyocytes with aging suggest that this cell population is an important determinant of the cardiac senescent phenotype. Collectively, the delayed electrical repolarization of aging cardiomyocytes may be viewed as a critical variable of the aging myopathy and its propensity to evolve into ventricular decompensation under stressful conditions.


Subject(s)
Action Potentials , Aging/physiology , Myocytes, Cardiac/physiology , Ventricular Function , Animals , Dogs , Female , Hemodynamics , Male
5.
Circ Res ; 114(1): 41-55, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24170267

ABSTRACT

RATIONALE: Hypoxia favors stem cell quiescence, whereas normoxia is required for stem cell activation, but whether cardiac stem cell (CSC) function is regulated by the hypoxic/normoxic state of the cell is currently unknown. OBJECTIVE: A balance between hypoxic and normoxic CSCs may be present in the young heart, although this homeostatic control may be disrupted with aging. Defects in tissue oxygenation occur in the old myocardium, and this phenomenon may expand the pool of hypoxic CSCs, which are no longer involved in myocyte renewal. METHODS AND RESULTS: Here, we show that the senescent heart is characterized by an increased number of quiescent CSCs with intact telomeres that cannot re-enter the cell cycle and form a differentiated progeny. Conversely, myocyte replacement is controlled only by frequently dividing CSCs with shortened telomeres; these CSCs generate a myocyte population that is chronologically young but phenotypically old. Telomere dysfunction dictates their actual age and mechanical behavior. However, the residual subset of quiescent young CSCs can be stimulated in situ by stem cell factor reversing the aging myopathy. CONCLUSIONS: Our findings support the notion that strategies targeting CSC activation and growth interfere with the manifestations of myocardial aging in an animal model. Although caution has to be exercised in the translation of animal studies to human beings, our data strongly suggest that a pool of functionally competent CSCs persists in the senescent heart and that this stem cell compartment can promote myocyte regeneration effectively, partly correcting the aging myopathy.


Subject(s)
Aging/drug effects , Cardiomyopathies/metabolism , Hypoxia/metabolism , Myoblasts, Cardiac/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Stem Cell Factor/pharmacology , Stem Cell Niche , Aging/metabolism , Animals , Cardiomyopathies/drug therapy , Cardiomyopathies/pathology , Cell Cycle , Cell Lineage , Cell Proliferation , Cellular Senescence/drug effects , Hypoxia/pathology , Mice , Mice, Inbred C57BL , Myoblasts, Cardiac/drug effects , Myoblasts, Cardiac/physiology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Stem Cell Factor/therapeutic use , Telomere Homeostasis
6.
Circulation ; 129(2): 157-72, 2014 Jan 14.
Article in English | MEDLINE | ID: mdl-24249720

ABSTRACT

BACKGROUND: The efficacy of bypass surgery in patients with ischemic cardiomyopathy is not easily predictable; preoperative clinical conditions may be similar, but the outcome may differ significantly. We hypothesized that the growth reserve of cardiac stem cells (CSCs) and circulating cytokines promoting CSC activation are critical determinants of ventricular remodeling in this patient population. METHODS AND RESULTS: To document the growth kinetics of CSCs, population-doubling time, telomere length, telomerase activity, and insulin-like growth factor-1 receptor expression were measured in CSCs isolated from 38 patients undergoing bypass surgery. Additionally, the blood levels of insulin-like growth factor-1, hepatocyte growth factor, and vascular endothelial growth factor were evaluated. The variables of CSC growth were expressed as a function of the changes in wall thickness, chamber diameter and volume, ventricular mass-to-chamber volume ratio, and ejection fraction, before and 12 months after surgery. A high correlation was found between indices of CSC function and cardiac anatomy. Negative ventricular remodeling was not observed if CSCs retained a significant growth reserve. The high concentration of insulin-like growth factor-1 systemically pointed to the insulin-like growth factor-1-insulin-like growth factor-1 receptor system as a major player in the adaptive response of the myocardium. hepatocyte growth factor, a mediator of CSC migration, was also high in these patients preoperatively, as was vascular endothelial growth factor, possibly reflecting the vascular growth needed before bypass surgery. Conversely, a decline in CSC growth was coupled with wall thinning, chamber dilation, and depressed ejection fraction. CONCLUSIONS: The telomere-telomerase axis, population-doubling time, and insulin-like growth factor-1 receptor expression in CSCs, together with a high circulating level of insulin-like growth factor-1, represent a novel biomarker able to predict the evolution of ischemic cardiomyopathy following revascularization.


Subject(s)
Coronary Artery Bypass , Myocardial Ischemia/pathology , Myocardial Ischemia/surgery , Myocardium/pathology , Stem Cells/pathology , Aged , Biomarkers/blood , Cell Proliferation , Cells, Cultured , Cytokines/blood , Female , Follow-Up Studies , Hepatocyte Growth Factor/blood , Humans , Male , Middle Aged , Myocardial Ischemia/blood , Predictive Value of Tests , Receptor, IGF Type 1/blood , Stem Cells/ultrastructure , Telomerase/physiology , Telomere/ultrastructure , Treatment Outcome , Vascular Endothelial Growth Factor A/blood
7.
Circulation ; 128(20): 2211-23, 2013 Nov 12.
Article in English | MEDLINE | ID: mdl-24141256

ABSTRACT

BACKGROUND: Aging negatively impacts on the function of resident human cardiac progenitor cells (hCPCs). Effective regeneration of the injured heart requires mobilization of hCPCs to the sites of damage. In the young heart, signaling by the guidance receptor EphA2 in response to the ephrin A1 ligand promotes hCPC motility and improves cardiac recovery after infarction. METHODS AND RESULTS: We report that old hCPCs are characterized by cell-autonomous inhibition of their migratory ability ex vivo and impaired translocation in vivo in the damaged heart. EphA2 expression was not decreased in old hCPCs; however, the elevated level of reactive oxygen species in aged cells induced post-translational modifications of the EphA2 protein. EphA2 oxidation interfered with ephrin A1-stimulated receptor auto-phosphorylation, activation of Src family kinases, and caveolin-1-mediated internalization of the receptor. Cellular aging altered the EphA2 endocytic route, affecting the maturation of EphA2-containing endosomes and causing premature signal termination. Overexpression of functionally intact EphA2 in old hCPCs corrected the defects in endocytosis and downstream signaling, enhancing cell motility. Based on the ability of phenotypically young hCPCs to respond efficiently to ephrin A1, we developed a novel methodology for the prospective isolation of live hCPCs with preserved migratory capacity and growth reserve. CONCLUSIONS: Our data demonstrate that the ephrin A1/EphA2 pathway may serve as a target to facilitate trafficking of hCPCs in the senescent myocardium. Importantly, EphA2 receptor function can be implemented for the selection of hCPCs with high therapeutic potential, a clinically relevant strategy that does not require genetic manipulation of stem cells.


Subject(s)
Adult Stem Cells/physiology , Aging/physiology , Cell Movement/physiology , Myocardium/cytology , Receptor, EphA2/metabolism , Signal Transduction/physiology , Adult , Adult Stem Cells/cytology , Aged , Cells, Cultured , Endocytosis/physiology , Ephrin-A1/metabolism , Female , Humans , Male , Middle Aged , Receptor, EphA2/genetics , Regeneration/physiology , Transferrin/metabolism
8.
Circulation ; 128(12): 1286-97, 2013 Sep 17.
Article in English | MEDLINE | ID: mdl-23983250

ABSTRACT

BACKGROUND: Little is known about the function of inositol 1,4,5-trisphosphate receptors (IP3Rs) in the adult heart experimentally. Moreover, whether these Ca(2+) release channels are present and play a critical role in human cardiomyocytes remains to be defined. IP3Rs may be activated after Gαq-protein-coupled receptor stimulation, affecting Ca(2+) cycling, enhancing myocyte performance, and potentially favoring an increase in the incidence of arrhythmias. METHODS AND RESULTS: IP3R function was determined in human left ventricular myocytes, and this analysis was integrated with assays in mouse myocytes to identify the mechanisms by which IP3Rs influence the electric and mechanical properties of the myocardium. We report that IP3Rs are expressed and operative in human left ventricular myocytes. After Gαq-protein-coupled receptor activation, Ca(2+) mobilized from the sarcoplasmic reticulum via IP3Rs contributes to the decrease in resting membrane potential, prolongation of the action potential, and occurrence of early afterdepolarizations. Ca(2+) transient amplitude and cell shortening are enhanced, and extrasystolic and dysregulated Ca(2+) elevations and contractions become apparent. These alterations in the electromechanical behavior of human cardiomyocytes are coupled with increased isometric twitch of the myocardium and arrhythmic events, suggesting that Gαq-protein-coupled receptor activation provides inotropic reserve, which is hampered by electric instability and contractile abnormalities. Additionally, our findings support the notion that increases in Ca(2+) load by IP3Rs promote Ca(2+) extrusion by forward-mode Na(+)/Ca(2+) exchange, an important mechanism of arrhythmic events. CONCLUSIONS: The Gαq-protein/coupled receptor/IP3R axis modulates the electromechanical properties of the human myocardium and its propensity to develop arrhythmias.


Subject(s)
Action Potentials/physiology , Calcium Signaling/physiology , Heart Failure/physiopathology , Inositol 1,4,5-Trisphosphate Receptors/physiology , Myocytes, Cardiac/physiology , Adult , Animals , Arrhythmias, Cardiac/physiopathology , Cells, Cultured , Female , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Heart Failure/genetics , Heart Ventricles/cytology , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Myocardial Contraction/physiology , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/physiology , Sarcoplasmic Reticulum/physiology , Signal Transduction/physiology
9.
N Engl J Med ; 364(19): 1795-806, 2011 May 12.
Article in English | MEDLINE | ID: mdl-21561345

ABSTRACT

BACKGROUND: Although progenitor cells have been described in distinct anatomical regions of the lung, description of resident stem cells has remained elusive. METHODS: Surgical lung-tissue specimens were studied in situ to identify and characterize human lung stem cells. We defined their phenotype and functional properties in vitro and in vivo. RESULTS: Human lungs contain undifferentiated human lung stem cells nested in niches in the distal airways. These cells are self-renewing, clonogenic, and multipotent in vitro. After injection into damaged mouse lung in vivo, human lung stem cells form human bronchioles, alveoli, and pulmonary vessels integrated structurally and functionally with the damaged organ. The formation of a chimeric lung was confirmed by detection of human transcripts for epithelial and vascular genes. In addition, the self-renewal and long-term proliferation of human lung stem cells was shown in serial-transplantation assays. CONCLUSIONS: Human lungs contain identifiable stem cells. In animal models, these cells participate in tissue homeostasis and regeneration. They have the undemonstrated potential to promote tissue restoration in patients with lung disease. (Funded by the National Institutes of Health.).


Subject(s)
Lung/cytology , Stem Cells/physiology , Adult , Animals , Clone Cells , Female , Humans , Lung/embryology , Lung/physiology , Mice , Mice, Inbred C57BL , Pluripotent Stem Cells , Proto-Oncogene Proteins c-kit/analysis , Regeneration , Stem Cell Transplantation , Stem Cells/chemistry
10.
Circ Res ; 108(9): 1071-83, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21415392

ABSTRACT

RATIONALE: Understanding the mechanisms that regulate trafficking of human cardiac stem cells (hCSCs) may lead to development of new therapeutic approaches for the failing heart. OBJECTIVE: We tested whether the motility of hCSCs in immunosuppressed infarcted animals is controlled by the guidance system that involves the interaction of Eph receptors with ephrin ligands. METHODS AND RESULTS: Within the cardiac niches, cardiomyocytes expressed preferentially the ephrin A1 ligand, whereas hCSCs possessed the EphA2 receptor. Treatment of hCSCs with ephrin A1 resulted in the rapid internalization of the ephrin A1-EphA2 complex, posttranslational modifications of Src kinases, and morphological changes consistent with the acquisition of a motile cell phenotype. Ephrin A1 enhanced the motility of hCSCs in vitro, and their migration in vivo following acute myocardial infarction. At 2 weeks after infarction, the volume of the regenerated myocardium was 2-fold larger in animals injected with ephrin A1-activated hCSCs than in animals receiving control hCSCs; this difference was dictated by a greater number of newly formed cardiomyocytes and coronary vessels. The increased recovery in myocardial mass with ephrin A1-treated hCSCs was characterized by further restoration of cardiac function and by a reduction in arrhythmic events. CONCLUSIONS: Ephrin A1 promotes the motility of EphA2-positive hCSCs, facilitates their migration to the area of damage, and enhances cardiac repair. Thus, in situ stimulation of resident hCSCs with ephrin A1 or their ex vivo activation before myocardial delivery improves cell targeting to sites of injury, possibly providing a novel strategy for the management of the diseased heart.


Subject(s)
Ephrin-A1/genetics , Ephrin-A2/genetics , Hematopoietic Stem Cells/cytology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/cytology , Animals , Cell Adhesion/physiology , Cell Membrane/metabolism , Cell Movement/physiology , Cytoplasm/metabolism , Ephrin-A1/metabolism , Ephrin-A2/metabolism , Gene Expression/physiology , Green Fluorescent Proteins/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Rats , Rats, Inbred F344 , Tachycardia, Ventricular/pathology , Tachycardia, Ventricular/physiopathology , Tachycardia, Ventricular/therapy
11.
Circ Res ; 108(7): 857-61, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21330601

ABSTRACT

RATIONALE: Two categories of cardiac stem cells (CSCs) with predominantly myogenic (mCSC) and vasculogenic (vCSC) properties have been characterized in the human heart. However, it is unknown whether functionally competent CSCs of both classes are present in the myocardium of patients affected by end-stage cardiac failure, and whether these cells can be harvested from relatively small myocardial samples. OBJECTIVE: To establish whether a clinically relevant number of mCSCs and vCSCs can be isolated and expanded from endomyocardial biopsies of patients undergoing cardiac transplantation or left ventricular assist device implantation. METHODS AND RESULTS: Endomyocardial biopsies were collected with a bioptome from the right side of the septum of explanted hearts or the apical LV core at the time of left ventricular assist device implantation. Two to 5 biopsies from each patient were enzymatically dissociated, and, after expansion, cells were sorted for c-kit (mCSCs) or c-kit and KDR (vCSCs) and characterized. mCSCs and vCSCs constituted 97% and 3% of the c-kit population, respectively. Population doubling time averaged 27 hours in mCSCs and vCSCs; 5×10(6) mCSCs and vCSCs were obtained in 28 and 41 days, respectively. Both CSC classes possessed significant growth reserve as documented by high telomerase activity and relatively long telomeres. mCSCs formed mostly cardiomyocytes, and vCSCs endothelial and smooth muscle cells. CONCLUSIONS: The growth properties of mCSCs and vCSCs isolated from endomyocardial biopsies from patients with advanced heart failure were comparable to those obtained previously from larger myocardial samples of patients undergoing elective cardiac surgery.


Subject(s)
Adult Stem Cells/pathology , Adult Stem Cells/physiology , Cardiomyopathies/pathology , Myocardium/pathology , Adult , Aged , Biopsy , Cardiomyopathies/physiopathology , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Female , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Male , Middle Aged , Telomere/pathology
12.
Circ Res ; 108(12): 1467-81, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21546606

ABSTRACT

RATIONALE: Age and coronary artery disease may negatively affect the function of human cardiac stem cells (hCSCs) and their potential therapeutic efficacy for autologous cell transplantation in the failing heart. OBJECTIVE: Insulin-like growth factor (IGF)-1, IGF-2, and angiotensin II (Ang II), as well as their receptors, IGF-1R, IGF-2R, and AT1R, were characterized in c-kit(+) hCSCs to establish whether these systems would allow us to separate hCSC classes with different growth reserve in the aging and diseased myocardium. METHODS AND RESULTS: C-kit(+) hCSCs were collected from myocardial samples obtained from 24 patients, 48 to 86 years of age, undergoing elective cardiac surgery for coronary artery disease. The expression of IGF-1R in hCSCs recognized a young cell phenotype defined by long telomeres, high telomerase activity, enhanced cell proliferation, and attenuated apoptosis. In addition to IGF-1, IGF-1R(+) hCSCs secreted IGF-2 that promoted myocyte differentiation. Conversely, the presence of IGF-2R and AT1R, in the absence of IGF-1R, identified senescent hCSCs with impaired growth reserve and increased susceptibility to apoptosis. The ability of IGF-1R(+) hCSCs to regenerate infarcted myocardium was then compared with that of unselected c-kit(+) hCSCs. IGF-1R(+) hCSCs improved cardiomyogenesis and vasculogenesis. Pretreatment of IGF-1R(+) hCSCs with IGF-2 resulted in the formation of more mature myocytes and superior recovery of ventricular structure. CONCLUSIONS: hCSCs expressing only IGF-1R synthesize both IGF-1 and IGF-2, which are potent modulators of stem cell replication, commitment to the myocyte lineage, and myocyte differentiation, which points to this hCSC subset as the ideal candidate cell for the management of human heart failure.


Subject(s)
Coronary Artery Disease/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Receptor, IGF Type 1/metabolism , Regeneration , Stem Cells/metabolism , Angiotensin II/metabolism , Cell Differentiation , Coronary Artery Disease/pathology , Coronary Artery Disease/therapy , Female , Humans , Insulin-Like Growth Factor I/biosynthesis , Insulin-Like Growth Factor II/metabolism , Male , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Myocardium/pathology , Myocytes, Cardiac/pathology , Receptor, IGF Type 2/metabolism , Stem Cell Transplantation , Stem Cells/pathology , Transplantation, Autologous
13.
J Immunol ; 187(4): 2006-14, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21753148

ABSTRACT

We previously demonstrated that anti-third-party CTLs (stimulated under IL-2 deprivation against cells with an MHC class I [MHC-I] background different from that of the host and the donor) are depleted of graft-versus-host reactivity and can eradicate B cell chronic lymphocytic leukemia cells in vitro or in an HU/SCID mouse model. We demonstrated in the current study that human allogeneic or autologous anti-third-party CTLs can also efficiently eradicate primary non-Hodgkin B cell lymphoma by inducing slow apoptosis of the pathological cells. Using MHC-I mutant cell line as target cells, which are unrecognizable by the CTL TCR, we demonstrated directly that this killing is TCR independent. Strikingly, this unique TCR-independent killing is induced through lymphoma MHC-I engagement. We further showed that this killing mechanism begins with durable conjugate formation between the CTLs and the tumor cells, through rapid binding of tumor ICAM-1 to the CTL LFA-1 molecule. This conjugation is followed by a slower second step of MHC-I-dependent apoptosis, requiring the binding of the MHC-I α2/3 C region on tumor cells to the CTL CD8 molecule for killing to ensue. By comparing CTL-mediated killing of Daudi lymphoma cells (lacking surface MHC-I expression) to Daudi cells with reconstituted surface MHC-I, we demonstrated directly for the first time to our knowledge, in vitro and in vivo, a novel role for MHC-I in the induction of lymphoma cell apoptosis by CTLs. Additionally, by using different knockout and transgenic strains, we further showed that mouse anti-third-party CTLs also kill lymphoma cells using similar unique TCR-independence mechanism as human CTLs, while sparing normal naive B cells.


Subject(s)
Apoptosis/immunology , CD8 Antigens/immunology , Histocompatibility Antigens Class I/immunology , Immunity, Cellular , Lymphoma, B-Cell/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Apoptosis/genetics , B-Lymphocytes/immunology , B-Lymphocytes/pathology , CD8 Antigens/genetics , Cell Line, Tumor , Female , Histocompatibility Antigens Class I/genetics , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/immunology , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, SCID , Mutation , T-Lymphocytes, Cytotoxic/pathology
14.
Nat Med ; 12(6): 657-64, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16715089

ABSTRACT

Here we investigated the potential role of bone-resorbing osteoclasts in homeostasis and stress-induced mobilization of hematopoietic progenitors. Different stress situations induced activity of osteoclasts (OCLs) along the stem cell-rich endosteum region of bone, secretion of proteolytic enzymes and mobilization of progenitors. Specific stimulation of OCLs with RANKL recruited mainly immature progenitors to the circulation in a CXCR4- and MMP-9-dependent manner; however, RANKL did not induce mobilization in young female PTPepsilon-knockout mice with defective OCL bone adhesion and resorption. Inhibition of OCLs with calcitonin reduced progenitor egress in homeostasis, G-CSF mobilization and stress situations. RANKL-stimulated bone-resorbing OCLs also reduced the stem cell niche components SDF-1, stem cell factor (SCF) and osteopontin along the endosteum, which was associated with progenitor mobilization. Finally, the major bone-resorbing proteinase, cathepsin K, also cleaved SDF-1 and SCF. Our findings indicate involvement of OCLs in selective progenitor recruitment as part of homeostasis and host defense, linking bone remodeling with regulation of hematopoiesis.


Subject(s)
Bone Resorption , Bone and Bones/anatomy & histology , Cell Movement/physiology , Hematopoietic Stem Cells/physiology , Osteoclasts/metabolism , Animals , Carrier Proteins/metabolism , Cathepsin K , Cathepsins/genetics , Cathepsins/metabolism , Cell Line , Chemokine CXCL12 , Chemokines, CXC/metabolism , Female , Hematopoietic Stem Cells/cytology , Homeostasis , Humans , Matrix Metalloproteinase 9/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred Strains , Mice, Knockout , Osteoclasts/cytology , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Receptor-Like Protein Tyrosine Phosphatases, Class 4 , Receptors, CXCR4/metabolism , Stem Cell Factor/metabolism
15.
Lancet ; 378(9806): 1847-57, 2011 11 26.
Article in English | MEDLINE | ID: mdl-22088800

ABSTRACT

BACKGROUND: c-kit-positive, lineage-negative cardiac stem cells (CSCs) improve post-infarction left ventricular (LV) dysfunction when administered to animals. We undertook a phase 1 trial (Stem Cell Infusion in Patients with Ischemic cardiOmyopathy [SCIPIO]) of autologous CSCs for the treatment of heart failure resulting from ischaemic heart disease. METHODS: In stage A of the SCIPIO trial, patients with post-infarction LV dysfunction (ejection fraction [EF] ≤40%) before coronary artery bypass grafting were consecutively enrolled in the treatment and control groups. In stage B, patients were randomly assigned to the treatment or control group in a 2:3 ratio by use of a computer-generated block randomisation scheme. 1 million autologous CSCs were administered by intracoronary infusion at a mean of 113 days (SE 4) after surgery; controls were not given any treatment. Although the study was open label, the echocardiographic analyses were masked to group assignment. The primary endpoint was short-term safety of CSCs and the secondary endpoint was efficacy. A per-protocol analysis was used. This study is registered with ClinicalTrials.gov, number NCT00474461. FINDINGS: This study is still in progress. 16 patients were assigned to the treatment group and seven to the control group; no CSC-related adverse effects were reported. In 14 CSC-treated patients who were analysed, LVEF increased from 30·3% (SE 1·9) before CSC infusion to 38·5% (2·8) at 4 months after infusion (p=0·001). By contrast, in seven control patients, during the corresponding time interval, LVEF did not change (30·1% [2·4] at 4 months after CABG vs 30·2% [2·5] at 8 months after CABG). Importantly, the salubrious effects of CSCs were even more pronounced at 1 year in eight patients (eg, LVEF increased by 12·3 ejection fraction units [2·1] vs baseline, p=0·0007). In the seven treated patients in whom cardiac MRI could be done, infarct size decreased from 32·6 g (6·3) by 7·8 g (1·7; 24%) at 4 months (p=0·004) and 9·8 g (3·5; 30%) at 1 year (p=0·04). INTERPRETATION: These initial results in patients are very encouraging. They suggest that intracoronary infusion of autologous CSCs is effective in improving LV systolic function and reducing infarct size in patients with heart failure after myocardial infarction, and warrant further, larger, phase 2 studies. FUNDING: University of Louisville Research Foundation and National Institutes of Health.


Subject(s)
Coronary Vessels , Myocardial Infarction/mortality , Myocardial Infarction/therapy , Stem Cell Transplantation/methods , Combined Modality Therapy , Coronary Artery Bypass/methods , Echocardiography, Doppler/methods , Female , Follow-Up Studies , Heart Failure/prevention & control , Heart Failure/therapy , Humans , Injections, Intra-Arterial , Magnetic Resonance Imaging/methods , Male , Middle Aged , Myocardial Infarction/diagnosis , Myocardial Ischemia/diagnosis , Myocardial Ischemia/mortality , Myocardial Ischemia/therapy , Myocytes, Cardiac/transplantation , Postoperative Care/methods , Prospective Studies , Reference Values , Risk Assessment , Survival Analysis , Time Factors , Tissue and Organ Harvesting , Transplantation, Autologous/methods , Treatment Outcome , Ventricular Remodeling/physiology
16.
Circ Res ; 107(11): 1374-86, 2010 Nov 26.
Article in English | MEDLINE | ID: mdl-21088285

ABSTRACT

RATIONALE: The turnover of cardiomyocytes in the aging female and male heart is currently unknown, emphasizing the need to define human myocardial biology. OBJECTIVE: The effects of age and gender on the magnitude of myocyte regeneration and the origin of newly formed cardiomyocytes were determined. METHODS AND RESULTS: The interaction of myocyte replacement, cellular senescence, growth inhibition, and apoptosis was measured in normal female (n=32) and male (n=42) human hearts collected from patients 19 to 104 years of age who died from causes other than cardiovascular diseases. A progressive loss of telomeric DNA in human cardiac stem cells (hCSCs) occurs with aging and the newly formed cardiomyocytes inherit short telomeres and rapidly reach the senescent phenotype. Our data provide novel information on the superior ability of the female heart to sustain the multiple variables associated with the development of the senescent myopathy. At all ages, the female heart is equipped with a larger pool of functionally competent hCSCs and younger myocytes than the male myocardium. The replicative potential is higher and telomeres are longer in female hCSCs than in male hCSCs. In the female heart, myocyte turnover occurs at a rate of 10%, 14%, and 40% per year at 20, 60, and 100 years of age, respectively. Corresponding values in the male heart are 7%, 12%, and 32% per year, documenting that cardiomyogenesis involves a large and progressively increasing number of parenchymal cells with aging. From 20 to 100 years of age, the myocyte compartment is replaced 15 times in women and 11 times in men. CONCLUSIONS: The human heart is a highly dynamic organ regulated by a pool of resident hCSCs that modulate cardiac homeostasis and condition organ aging.


Subject(s)
Adult Stem Cells/cytology , Adult Stem Cells/physiology , Cell Differentiation/physiology , Cellular Senescence/physiology , Heart/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Adult , Aged , Aged, 80 and over , Apoptosis/physiology , Cell Death/physiology , Cells, Cultured , Female , Heart/anatomy & histology , Humans , Male , Middle Aged , Sex Characteristics , Young Adult
17.
Circ Res ; 107(2): 305-15, 2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20522802

ABSTRACT

RATIONALE: The ability of the human heart to regenerate large quantities of myocytes remains controversial, and the extent of myocyte renewal claimed by different laboratories varies from none to nearly 20% per year. OBJECTIVE: To address this issue, we examined the percentage of myocytes, endothelial cells, and fibroblasts labeled by iododeoxyuridine in postmortem samples obtained from cancer patients who received the thymidine analog for therapeutic purposes. Additionally, the potential contribution of DNA repair, polyploidy, and cell fusion to the measurement of myocyte regeneration was determined. METHODS AND RESULTS: The fraction of myocytes labeled by iododeoxyuridine ranged from 2.5% to 46%, and similar values were found in fibroblasts and endothelial cells. An average 22%, 20%, and 13% new myocytes, fibroblasts, and endothelial cells were generated per year, suggesting that the lifespan of these cells was approximately 4.5, 5, and 8 years, respectively. The newly formed cardiac cells showed a fully differentiated adult phenotype and did not express the senescence-associated protein p16(INK4a). Moreover, measurements by confocal microscopy and flow cytometry documented that the human heart is composed predominantly of myocytes with 2n diploid DNA content and that tetraploid and octaploid nuclei constitute only a small fraction of the parenchymal cell pool. Importantly, DNA repair, ploidy formation, and cell fusion were not implicated in the assessment of myocyte regeneration. CONCLUSIONS: Our findings indicate that the human heart has a significant growth reserve and replaces its myocyte and nonmyocyte compartment several times during the course of life.


Subject(s)
Cell Proliferation , Endothelial Cells/pathology , Fibroblasts/pathology , Muscle Development , Myocardium/pathology , Myocytes, Cardiac/pathology , Neoplasms/pathology , Adult , Age Factors , Aged , Animals , Autopsy , Cell Death , Cell Fusion , Cell Nucleus/pathology , Cell Proliferation/drug effects , DNA Repair , Endothelial Cells/drug effects , Female , Fibroblasts/drug effects , Flow Cytometry , Humans , Idoxuridine/therapeutic use , Immunohistochemistry , Male , Microscopy, Confocal , Middle Aged , Muscle Development/drug effects , Myocytes, Cardiac/drug effects , Neoplasms/drug therapy , Phenotype , Polyploidy , Radiation-Sensitizing Agents/therapeutic use , Rats , Rats, Inbred F344 , Regeneration , Time Factors , Young Adult
20.
J Clin Invest ; 115(1): 168-76, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15630457

ABSTRACT

The chemokine stromal cell-derived factor-1 (SDF-1) and its receptor, CXCR4, play a major role in migration, retention, and development of hematopoietic progenitors in the bone marrow. We report the direct involvement of atypical PKC-zeta in SDF-1 signaling in immature human CD34(+)-enriched cells and in leukemic pre-B acute lymphocytic leukemia (ALL) G2 cells. Chemotaxis, cell polarization, and adhesion of CD34(+) cells to bone marrow stromal cells were found to be PKC-zeta dependent. Overexpression of PKC-zeta in G2 and U937 cells led to increased directional motility to SDF-1. Interestingly, impaired SDF-1-induced migration of the pre-B ALL cell line B1 correlated with reduced PKC-zeta expression. SDF-1 triggered PKC-zeta phosphorylation, translocation to the plasma membrane, and kinase activity. Furthermore we identified PI3K as an activator of PKC-zeta, and Pyk-2 and ERK1/2 as downstream targets of PKC-zeta. SDF-1-induced proliferation and MMP-9 secretion also required PKC-zeta activation. Finally, we showed that in vivo engraftment, but not homing, of human CD34(+)-enriched cells to the bone marrow of NOD/SCID mice was PKC-zeta dependent and that injection of mice with inhibitory PKC-zeta pseudosubstrate peptides resulted in mobilization of murine progenitors. Our results demonstrate a central role for PKC-zeta in SDF-1-dependent regulation of hematopoietic stem and progenitor cell motility and development.


Subject(s)
Antigens, CD34/metabolism , Cell Differentiation , Cell Movement , Chemokines, CXC/metabolism , Protein Kinase C/metabolism , Stem Cells/cytology , Stem Cells/enzymology , Actins/metabolism , Animals , Burkitt Lymphoma/metabolism , Burkitt Lymphoma/pathology , Cell Adhesion/drug effects , Cell Movement/drug effects , Cells, Cultured , Chemokine CXCL12 , Chemokines, CXC/pharmacology , Enzyme Activation , Fetal Blood/cytology , Fetal Blood/drug effects , Fetal Blood/metabolism , Humans , Mice , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Stem Cells/drug effects , Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL