Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Acta Biomater ; 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38942187

ABSTRACT

Applied to the epicardium in-vivo, regenerative cardiac patches support the ventricular wall, reduce wall stresses, encourage ventricular wall thickening, and improve ventricular function. Scaffold engraftment, however, remains a challenge. After implantation, scaffolds are subject to the complex, time-varying, biomechanical environment of the myocardium. The mechanical capacity of engineered tissue to biomimetically deform and simultaneously support the damaged native tissue is crucial for its efficacy. To date, however, the biomechanical response of engineered tissue applied directly to live myocardium has not been characterized. In this paper, we utilize optical imaging of a Langendorff ex-vivo cardiac model to characterize the native deformation of the epicardium as well as that of attached engineered scaffolds. We utilize digital image correlation, linear strain, and 2D principal strain analysis to assess the mechanical compliance of acellular ice templated collagen scaffolds. Scaffolds had either aligned or isotropic porous architecture and were adhered directly to the live epicardial surface with either sutures or cyanoacrylate glue. We demonstrate that the biomechanical characteristics of native myocardial deformation on the epicardial surface can be reproduced by an ex-vivo cardiac model. Furthermore, we identified that scaffolds with unidirectionally aligned pores adhered with suture fixation most accurately recapitulated the deformation of the native epicardium. Our study contributes a translational characterization methodology to assess the physio-mechanical performance of engineered cardiac tissue and adds to the growing body of evidence showing that anisotropic scaffold architecture improves the functional biomimetic capacity of engineered cardiac tissue. STATEMENT OF SIGNIFICANCE: Engineered cardiac tissue offers potential for myocardial repair, but engraftment remains a challenge. In-vivo, engineered scaffolds are subject to complex biomechanical stresses and the mechanical capacity of scaffolds to biomimetically deform is critical. To date, the biomechanical response of engineered scaffolds applied to live myocardium has not been characterized. In this paper, we utilize optical imaging of an ex-vivo cardiac model to characterize the deformation of the native epicardium and scaffolds attached directly to the heart. Comparing scaffold architecture and fixation method, we demonstrate that sutured scaffolds with anisotropic pores aligned with the native alignment of the superficial myocardium best recapitulate native deformation. Our study contributes a physio-mechanical characterization methodology for cardiac tissue engineering scaffolds.

2.
PLoS One ; 7(8): e42527, 2012.
Article in English | MEDLINE | ID: mdl-22880017

ABSTRACT

Embryonic heart valve primordia (cushions) maintain unidirectional blood flow during development despite an increasingly demanding mechanical environment. Recent studies demonstrate that atrioventricular (AV) cushions stiffen over gestation, but the molecular mechanisms of this process are unknown. Transforming growth factor-beta (TGFß) and serotonin (5-HT) signaling modulate tissue biomechanics of postnatal valves, but less is known of their role in the biomechanical remodeling of embryonic valves. In this study, we demonstrate that exogenous TGFß3 increases AV cushion biomechanical stiffness and residual stress, but paradoxically reduces matrix compaction. We then show that TGFß3 induces contractile gene expression (RhoA, aSMA) and extracellular matrix expression (col1α2) in cushion mesenchyme, while simultaneously stimulating a two-fold increase in proliferation. Local compaction increased due to an elevated contractile phenotype, but global compaction appeared reduced due to proliferation and ECM synthesis. Blockade of TGFß type I receptors via SB431542 inhibited the TGFß3 effects. We next showed that exogenous 5-HT does not influence cushion stiffness by itself, but synergistically increases cushion stiffness with TGFß3 co-treatment. 5-HT increased TGFß3 gene expression and also potentiated TGFß3 induced gene expression in a dose-dependent manner. Blockade of the 5HT2b receptor, but not 5-HT2a receptor or serotonin transporter (SERT), resulted in complete cessation of TGFß3 induced mechanical strengthening. Finally, systemic 5-HT administration in ovo induced cushion remodeling related defects, including thinned/atretic AV valves, ventricular septal defects, and outflow rotation defects. Elevated 5-HT in ovo resulted in elevated remodeling gene expression and increased TGFß signaling activity, supporting our ex-vivo findings. Collectively, these results highlight TGFß/5-HT signaling as a potent mechanism for control of biomechanical remodeling of AV cushions during development.


Subject(s)
Heart Valves/embryology , Heart Valves/physiology , Serotonin/pharmacology , Transforming Growth Factor beta3/pharmacology , Ventricular Remodeling/drug effects , Animals , Biomechanical Phenomena/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chick Embryo , Gene Expression Regulation, Developmental/drug effects , Heart Septal Defects/embryology , Heart Septal Defects/pathology , Heart Septal Defects/physiopathology , Heart Valves/drug effects , Heart Valves/pathology , Humans , Mesoderm/drug effects , Myocardial Contraction/drug effects , Phenotype , Protein Serine-Threonine Kinases/metabolism , Receptor, Serotonin, 5-HT2B/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Smad Proteins/metabolism , Ventricular Remodeling/genetics
3.
Dev Dyn ; 240(6): 1322-34, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21538685

ABSTRACT

Regulation of organ growth is critical during embryogenesis. At the cellular level, mechanisms controlling the size of individual embryonic organs include cell proliferation, differentiation, migration, and attrition through cell death. All these mechanisms play a role in cardiac morphogenesis, but experimental studies have shown that the major determinant of cardiac size during prenatal development is myocyte proliferation. As this proliferative capacity becomes severely restricted after birth, the number of cell divisions that occur during embryogenesis limits the growth potential of the postnatal heart. We summarize here current knowledge concerning regional control of myocyte proliferation as related to cardiac morphogenesis and dysmorphogenesis. There are significant spatial and temporal differences in rates of cell division, peaking during the preseptation period and then gradually decreasing toward birth. Analysis of regional rates of proliferation helps to explain the mechanics of ventricular septation, chamber morphogenesis, and the development of the cardiac conduction system. Proliferation rates are influenced by hemodynamic loading, and transduced by autocrine and paracrine signaling by means of growth factors. Understanding the biological response of the developing heart to such factors and physical forces will further our progress in engineering artificial myocardial tissues for heart repair and designing optimal treatment strategies for congenital heart disease.


Subject(s)
Cell Proliferation , Heart/embryology , Myocytes, Cardiac/physiology , Animals , Cell Differentiation/genetics , Gene Expression Regulation, Developmental , Heart/growth & development , Humans , Models, Biological , Morphogenesis/genetics , Morphogenesis/physiology , Myocardium/metabolism , Myocytes, Cardiac/metabolism
5.
Dev Dyn ; 238(6): 1535-46, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19418446

ABSTRACT

The hypothesis that inner layers of contracting muscular tubes undergo greater strain than concentric outer layers was tested by numerical modeling and by confocal microscopy of strain within the wall of the early chick heart. We modeled the looped heart as a thin muscular shell surrounding an inner layer of sponge-like trabeculae by two methods: calculation within a two-dimensional three-variable lumped model and simulated expansion of a three-dimensional, four-layer mesh of finite elements. Analysis of both models, and correlative microscopy of chamber dimensions, sarcomere spacing, and membrane leaks, indicate a gradient of strain decreasing across the wall from highest strain along inner layers. Prediction of wall thickening during expansion was confirmed by ultrasonography of beating hearts. Degree of stretch determined by radial position may thus contribute to observed patterns of regional myocardial conditioning and slowed proliferation, as well as to the morphogenesis of ventricular trabeculae and conduction fascicles. Developmental Dynamics 238:1535-1546, 2009. (c) 2009 Wiley-Liss, Inc.


Subject(s)
Heart Ventricles , Heart , Models, Cardiovascular , Animals , Chick Embryo , Echocardiography , Heart/anatomy & histology , Heart/embryology , Heart Ventricles/anatomy & histology , Heart Ventricles/diagnostic imaging , Heart Ventricles/embryology , Myocardial Contraction/physiology , Sarcomeres/diagnostic imaging , Sarcomeres/metabolism , Stress, Mechanical
6.
Dev Biol ; 319(2): 336-45, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18538758

ABSTRACT

Here we report that mouse embryos homozygous for a gene trap insertion in the fibulin-1 (Fbln1) gene are deficient in Fbln1 and exhibit cardiac ventricular wall thinning and ventricular septal defects with double outlet right ventricle or overriding aorta. Fbln1 nulls also display anomalies of aortic arch arteries, hypoplasia of the thymus and thyroid, underdeveloped skull bones, malformations of cranial nerves and hemorrhagic blood vessels in the head and neck. The spectrum of malformations is consistent with Fbln1 influencing neural crest cell (NCC)-dependent development of these tissues. This is supported by evidence that Fbln1 expression is associated with streams of cranial NCCs migrating adjacent to rhombomeres 2-7 and that Fbln1-deficient embryos display patterning anomalies of NCCs forming cranial nerves IX and X, which derive from rhombomeres 6 and 7. Additionally, Fbln1-deficient embryos show increased apoptosis in areas populated by NCCs derived from rhombomeres 4, 6 and 7. Based on these findings, it is concluded that Fbln1 is required for the directed migration and survival of cranial NCCs contributing to the development of pharyngeal glands, craniofacial skeleton, cranial nerves, aortic arch arteries, cardiac outflow tract and cephalic blood vessels.


Subject(s)
Calcium-Binding Proteins/genetics , Morphogenesis/physiology , Neural Crest/physiology , Animals , CD4 Antigens/genetics , Calcium-Binding Proteins/deficiency , Cerebrovascular Circulation/genetics , Chromosome Mapping , Crosses, Genetic , Endoplasmic Reticulum/physiology , Fetal Heart/pathology , Fetal Heart/physiology , Genotype , Heart Ventricles/embryology , Heart Ventricles/pathology , Immunohistochemistry , Mice , Mice, Knockout , Polymerase Chain Reaction , beta-Galactosidase/genetics
7.
Dev Dyn ; 236(12): 3503-13, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17948299

ABSTRACT

The chick embryo has long been a favorite model system for morphologic and physiologic studies of the developing heart, largely because of its easy visualization and amenability to experimental manipulations. However, this advantage is diminished after 5 days of incubation, when rapidly growing chorioallantoic membranes reduce visibility of the embryo. Using high-frequency ultrasound, we show that chick embryonic cardiovascular structures can be readily visualized throughout the period of Stages 9-39. At most stages of development, a simple ex ovo culture technique provided the best imaging opportunities. We have measured cardiac and vascular structures, blood flow velocities, and calculated ventricular volumes as early as Stage 11 with values comparable to those previously obtained using video microscopy. The endocardial and myocardial layers of the pre-septated heart are readily seen as well as the acellular layer of the cardiac jelly. Ventricular inflow in the pre-septated heart is biphasic, just as in the mature heart, and is converted to a monophasic (outflow) wave by ventricular contraction. Although blood has soft-tissue density at the ultrasound resolutions and developmental stages examined, its movement allowed easy discrimination of perfused vascular structures throughout the embryo. The utility of such imaging was demonstrated by documenting changes in blood flow patterns after experimental conotruncal banding.


Subject(s)
Cardiovascular System/diagnostic imaging , Cardiovascular System/embryology , Animals , Chick Embryo , Echocardiography/methods , Heart/embryology , Hemodynamics , Time Factors
8.
Dev Dyn ; 236(3): 671-83, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17226818

ABSTRACT

An important phase of cardiac outflow tract (OFT) formation is the remodeling of the distal region of the common outlet in which the myocardial sleeve is replaced by with smooth muscle. Here we demonstrate that expression of the proteoglycan versican is reduced before the loss of myocardium from the distal cardiac outlet concomitant with an increase in production of the N-terminal cleavage fragment of versican. To test whether versican proteolysis plays a role in OFT remodeling, we determined the effects of adenoviral-mediated expression of a versican isoform devoid of known matrix metalloproteinase cleavage sites (V3) and an N-terminal fragment of versican (G1). V3 expression promoted an increase in thickness of the proximal OFT myocardial layer independent of proliferation. In contrast, the G1 domain caused thinning and interruptions of the OFT myocardium. These in vivo findings were consistent with findings using cultured primary cardiomyocytes showing that the V3 promoted myocardial cell-cell association while the G1 domain caused a loss of myocardial cell-cell association. Taken together, we conclude that intact versican and G1-containing versican cleavage products have opposing effects on myocardial cells and that versican proteolysis may facilitate the loss of distal myocardium during OFT remodeling.


Subject(s)
Embryonic Development/physiology , Heart/physiology , Myocardium/metabolism , Versicans/metabolism , Adenoviridae/genetics , Animals , Binding Sites/genetics , Chick Embryo , Chickens , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Heart/embryology , Hyaluronic Acid/metabolism , Immunohistochemistry , Matrix Metalloproteinases/metabolism , Mice , Microinjections , Microscopy, Confocal , Models, Genetic , Mutation , Myocardium/cytology , Versicans/genetics , Versicans/physiology
9.
Am J Physiol Heart Circ Physiol ; 291(4): H1646-52, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16679393

ABSTRACT

To characterize developmental changes in impulse propagation within atrial musculature, we performed high-speed optical mapping of activation sequence of the developing chick atria using voltage-sensitive dye. The activation maps were correlated with detailed morphological studies using scanning electron microscopy, histology, and whole mount confocal imaging with three-dimensional reconstruction. A preferential pathway appeared during development within the roof of the atria, transmitting the impulse rapidly from the right-sided sinoatrial node to the left atrium. The morphological substrate of this pathway, the bundle of Bachman, apparent from stage 29 onward, was a prominent ridge of pectinate muscles continuous with the terminal crest. Further acceleration of impulse propagation was noted along the ridges formed by the developing pectinate muscles, ramifying from the terminal crest toward the atrioventricular groove. In contrast, when the impulse reached the interatrial septum, slowing was often observed, suggesting that the septum acts as a barrier or sink for electrical current. We conclude that these inhomogeneities in atrial impulse propagation are consistent with existence of a specialized network of fast-conducting tissues. The purpose of these preferential pathways appears to be to assure synchronous atrial activation and contraction rather than rapid impulse conduction between the sinoatrial and atrioventricular nodes.


Subject(s)
Atrial Function/physiology , Heart Atria/embryology , Heart Conduction System/physiology , Myocardium/cytology , Animals , Atrioventricular Node/physiology , Chick Embryo , Electrophysiology , Heart Atria/cytology , Heart Conduction System/cytology , Myocardial Contraction/physiology , Sinoatrial Node/physiology
10.
Microsc Microanal ; 11(3): 209-15, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16060973

ABSTRACT

Specialized conduction tissues mediate coordinated propagation of electrical activity through the adult vertebrate heart. Following activation of the atria, the activation wave is slowed down in the atrioventricular canal or node, after which it spreads rapidly into the left and right ventricles via the His-Purkinje system (HPS). This results in the ventricles being activated from the apex toward the base, which is a hallmark of HPS function. The development of mature HPS function follows significant phases of cardiac morphogenesis. Initially, the cardiac impulse propagates in a slow, linear, and isotropic fashion from the sinus venosus at the most caudal portion of the tubular heart. Although the speed of impulse propagation gradually increases as it travels toward the anterior regions of the heart tube, the actual sequence of ventricular activation in the looped heart proceeds in the same direction as blood flow. Eventually, the immature base-to-apex sequence of ventricular activation undergoes an apparent reversal, changing to the mature apex-to-base pattern. Using an optical mapping approach, we demonstrate that the timing of this last transition shows striking dependence on hemodynamic loading of the ventricle, being accelerated by pressure overload and delayed in left ventricular hypoplasia. Comparison of chick and mammalian hearts revealed some striking similarities as well as key differences in the timing of such events during cardiac organogenesis.


Subject(s)
Heart/embryology , Action Potentials , Animals , Bundle of His/embryology , Female , Heart/physiology , Heart Ventricles/embryology , Models, Animal , Pregnancy , Rats
11.
Microsc Microanal ; 11(3): 216-23, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16060974

ABSTRACT

Confocal microscopy allows for optical sectioning of tissues, thus obviating the need for physical sectioning and subsequent registration to obtain a three-dimensional representation of tissue architecture. However, practicalities such as tissue opacity, light penetration, and detector sensitivity have usually limited the available depth of imaging to 200 microm. With the emergence of newer, more powerful systems, we attempted to push these limits to those dictated by the working distance of the objective. We used whole-mount immunohistochemical staining followed by clearing with benzyl alcohol-benzyl benzoate (BABB) to visualize three-dimensional myocardial architecture. Confocal imaging of entire chick embryonic hearts up to a depth of 1.5 mm with voxel dimensions of 3 microm was achieved with a 10x dry objective. For the purpose of screening for congenital heart defects, we used endocardial painting with fluorescently labeled poly-L-lysine and imaged BABB-cleared hearts with a 5x objective up to a depth of 2 mm. Two-photon imaging of whole-mount specimens stained with Hoechst nuclear dye produced clear images all the way through stage 29 hearts without significant signal attenuation. Thus, currently available systems allow confocal imaging of fixed samples to previously unattainable depths, the current limiting factors being objective working distance, antibody penetration, specimen autofluorescence, and incomplete clearing.


Subject(s)
Heart/embryology , Animals , Chick Embryo , Heart Defects, Congenital/pathology , Imaging, Three-Dimensional , Immunohistochemistry , Microscopy, Confocal
12.
Cardiovasc Res ; 65(4): 842-50, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15721864

ABSTRACT

OBJECTIVES: The aim of this study is to analyze Scn1b mRNA expression levels and protein distribution of Scn1b, a putative modulator of the pore-forming Na(+) channel subunit in the heart, during mouse cardiac development. METHODS: Scn1b mRNA levels were determined by real-time RT-PCR using embryonic hearts ranging from E9.5 to E18.5 as well as in postnatal and adult heart. Scn1b protein distribution and subcellular localization during cardiogenesis were analyzed by immunohistochemistry and confocal microscopy. RESULTS: Scn1b mRNA showed a dynamic expression pattern, peaking at stage E12.5 and decreasing at E15.5. Scn1b mRNA increased at later embryonic and neonatal stages, being maximal in the adult heart. Immunohistochemistry experiments revealed comparable distribution of Scn1b protein between the different cardiac chambers at early embryonic stages. With further development, Scn1b protein showed an enhanced expression in the trabeculated myocardium and the bundle branches. At the subcellular level in later embryonic and postnatal mouse cardiomyocytes, Scn1b was present in T-tubules as identified by immunostaining of alpha-actinin, and in the intercalated disks as identified by immunostaining of connexin 43. CONCLUSION: These results demonstrate that Scn1b is expressed during mouse heart development, suggesting it can play an important role in the action potential configuration of the cardiomyocytes during heart morphogenesis.


Subject(s)
Fetal Heart/metabolism , Sodium Channels/metabolism , Aging/metabolism , Animals , Embryonic Development , Female , Gene Expression Regulation, Developmental , Mice , Mice, Inbred BALB C , Purkinje Fibers/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Sodium Channels/genetics , Voltage-Gated Sodium Channel beta-1 Subunit
13.
Dev Dyn ; 231(4): 849-58, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15517585

ABSTRACT

To evaluate the developmental distribution of adrenergic cells in vivo, we inserted the Cre-recombinase gene into the locus encoding for the epinephrine biosynthetic enzyme phenylethanolamine n-methyltransferase (Pnmt) and crossed these Pnmt-Cre mice with ROSA26 reporter (R26R) mice to activate LacZ (encoding beta-galactosidase) expression in cells that were selectively derived from the adrenergic lineage. Our data show the following: (1) Insertion of Cre-recombinase into the Pnmt locus created a functional knockout of Pnmt expression with concomitant loss of epinephrine in homozygous Pnmt(Cre/Cre) mice; (2) Despite the reduction in Pnmt expression and epinephrine production in Pnmt(Cre/Cre) mice, these mice were viable and fertile, with no apparent developmental defects; (3) When crossed with R26R mice, Pnmt-Cre activation of LacZ expression faithfully recapitulated Pnmt expression in vivo; and (4) LacZ expression was activated in substantial numbers of pacemaking, conduction, and working cardiomyocytes.


Subject(s)
Adrenal Glands/embryology , Epinephrine/physiology , Integrases/genetics , Phenylethanolamine N-Methyltransferase/genetics , Adrenal Glands/cytology , Adrenal Glands/physiology , Animals , Female , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Genes, Reporter , Lac Operon , Male , Mice , Mice, Knockout , Models, Animal , Mutagenesis, Insertional , Pregnancy
14.
Anat Rec A Discov Mol Cell Evol Biol ; 280(2): 1001-9, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15368341

ABSTRACT

The specialized conduction tissue network mediates coordinated propagation of electrical activity through the adult vertebrate heart. Following activation of the atria, the activation wave is slowed down in the atrioventricular canal or node, then spreads rapidly into the left and right ventricles via the His-Purkinje system (HPS). This results in the ventricle being activated from the apex toward the base and is thought to represent HPS function. The development of mature HPS function in embryogenesis follows significant phases of cardiac morphogenesis. Initially, cardiac impulse propagates in a slow, linear, and isotropic fashion from the sinus venosus at the most caudal portion of the tubular heart. Although the speed of impulse propagation gradually increases, ventricular activation in the looped heart still follows the direction of blood flow. Eventually, the immature base-to-apex sequence of ventricular activation undergoes an apparent reversal, maturing to apex-to-base pattern. The embryonic chick heart has been studied intensively by both electrophysiological and morphological techniques, and the morphology of its conduction system (which is similar to mammals) is well characterized. One interesting but seldom studied feature is the anterior septal branch (ASB), which came sharply to focus (together with the rest of the ventricular conduction system) in our birthdating studies. Using an optical mapping approach, we show that ASB serves to activate ventricular surface between stages 16 and 25, predating the functionality of the His bundle/bundle branches. Heart morphogenesis and conduction system formation are thus linked, and studying the abnormal activation patterns could further our understanding of pathogenesis of congenital heart disease.


Subject(s)
Action Potentials , Bundle of His/embryology , Chick Embryo/physiology , Heart Conduction System , Heart Ventricles , Animals , Electric Stimulation , Electrophysiology , Heart Conduction System/embryology , Heart Conduction System/physiology , Heart Ventricles/embryology , Ventricular Function
15.
Circulation ; 110(11 Suppl 1): II268-73, 2004 Sep 14.
Article in English | MEDLINE | ID: mdl-15364874

ABSTRACT

OBJECTIVE: The cause of thoracic aortic aneurysms (TAAs) is poorly understood. Previous work has suggested an association between development of aortic aneurysms and matrix metalloproteinase (MMP) activity. We hypothesized that removal of the primary endogenous aortic MMP inhibitor (TIMP) through TIMP-1 gene deletion will increase TAA progression. METHODS AND RESULTS: The descending thoracic aortas of wild-type 129 SvE and TIMP-1 gene knockout (TIMP-1-/-) mice were exposed to 0.5 mol/L CaCl2 for 15 minutes, with terminal studies performed at 4 or 8 weeks. TAA lumen diameter was measured using confocal microscopy and normalized to the ascending aorta. In addition, sections were studied with in situ zymography and immunohistochemistry staining for MMP-9. Both wild-type [TAA/ascending ratio (mean+/-SEM): control, 0.85+/-0.02 (n=14); 4 weeks, 1.00+/-0.03 (n=13); 8 weeks, 1.05+/-0.10 (n=9)] and TIMP-1-/- [control, 0.98+/-0.04 (n=11); 4 weeks, 1.10+/-0.03 (n =21); 8 weeks, 1.22+/-0.09 (n=10)] groups developed aneurysms at 4 and 8 weeks compared with their respective controls (P<0.05). TIMP-1-/- animals developed larger aneurysms than the corresponding wild-type group (P<0.05). Aneurysms in the TIMP-1-/- group were larger at 8 weeks than at 4 weeks (P<0.05), which was not seen in the wild-type aneurysm groups. Both groups showed presence of MMP-9 in 4 and 8 weeks, most prominently in the adventitia and outer media. In situ zymographic activity was increased in the 8-week TIMP-1-/- group compared with wild-type. CONCLUSIONS: Deletion of the TIMP-1 gene results in increased and continued progression of aneurysm formation compared with wild-type mice in a unique TAA model caused at least in part by an alteration in the balance between gelatinase activity and its endogenous inhibition. Therapeutic strategies aimed at modifying MMP activity may reduce or prevent the progression of TAAs.


Subject(s)
Aortic Aneurysm, Thoracic/pathology , Tissue Inhibitor of Metalloproteinase-1/deficiency , Animals , Aortic Aneurysm, Thoracic/enzymology , Disease Models, Animal , Disease Progression , Female , Gene Deletion , Male , Matrix Metalloproteinase 9/analysis , Matrix Metalloproteinases/physiology , Mice , Mice, Knockout , Microscopy, Confocal , Phenotype , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/physiology
16.
J Surg Res ; 115(1): 157-63, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14572787

ABSTRACT

BACKGROUND: The mechanisms of thoracic aortic aneurysm (TAA) formation are poorly understood, mainly due to the lack of a useful and reproducible model. Accordingly, the goal of this study was to test the hypothesis that abluminal calcium chloride (CaCl(2)) application could create TAAs in the mouse. MATERIALS AND METHODS: Adult 129/SvE mice (n = 8) were anesthetized and their thoracic aortas exposed via left thoracotomy. CaCl(2) (0.5M) was applied to the distal descending thoracic aorta for 15 min followed by chest closure. At 4 weeks, the perfusion-fixed aorta was harvested from the root to the renal arteries. Diameter measurements were made using confocal microscopy, and wall thickness was measured from hematoxylin and eosin-stained sections. RESULTS: The control (n = 15) distal descending thoracic aortic diameter was 0.60 +/- 0.04 mm and increased by 25% (0.76 +/- 0.06 mm, P < 0.05) following CaCl(2) treatment. Control aortic wall thickness was 48 +/- 9 mum and decreased by 47% in corresponding CaCl(2)-exposed segments (25 +/- 8 mum, P < 0.05). The diameter and wall thickness of the ascending aorta (used as an internal control) were not significantly different between groups. Picrosirius red staining of the TAA showed adventitial collagen breakdown and disruption of lamellar organization. CONCLUSIONS: We conclude that abluminal application of CaCl(2) to the thoracic aorta reliably produces dilation, wall-thinning, and disruption of mural architecture, the hallmark signs of aneurysm formation. To our knowledge, these findings describe for the first time the generation of a reproducible model of isolated TAA formation in a murine system.


Subject(s)
Aortic Aneurysm, Thoracic/pathology , Disease Models, Animal , Animals , Aorta, Thoracic/chemistry , Aorta, Thoracic/pathology , Aortic Aneurysm, Thoracic/chemically induced , Calcium Chloride/administration & dosage , Collagen/analysis , Dilatation, Pathologic , Female , Male , Mice
17.
Novartis Found Symp ; 250: 110-22; discussion 122-4, 276-9, 2003.
Article in English | MEDLINE | ID: mdl-12956326

ABSTRACT

The heartbeat is initiated and coordinated by a multi-component set of specialized muscle tissues collectively referred to as the pacemaking and conduction system. Over the last few years, impetus has gathered into unravelling the cellular and molecular processes that regulate differentiation and integration of this essential cardiac network. One focus of our collective work has been the developmental history of cells comprising His-Purkinje tissues of the conduction system. This interest in part arose from studies of the expression of connexins in periarterial Purkinje fibres of the chick heart. Using lineage-tracing strategies, including those based on replication-defective retroviruses and adenoviruses, it has been shown that conduction cells are derived from multipotent, cardiomyogenic progenitors in the tubular heart. Moreover, heterogeneity within myocardial clones has indicated that the elaboration of the conduction system in the chick embryo occurs by progressive, localized recruitment from within this pool of cardiomyogenic cells. Cell birth dating has revealed that inductive conscription of cells to central elements of the conduction system (e.g. the His bundle) precedes recruitment to the peripheral components of the network (i.e. subendocardial and periarterial Purkinje fibres). Birth dating studies in rodents suggest an analogous recruitment process is occurring in this species. In addition to summarizing earlier work, this chapter provides information on ongoing studies of cell-cell signalling and transcriptional mechanisms that may regulate the development of His-Purkinje tissues.


Subject(s)
Bundle of His/growth & development , Cell Lineage , Heart/anatomy & histology , Myocardium , Purkinje Fibers/growth & development , Animals , Bundle of His/cytology , Cell Differentiation/physiology , Chick Embryo , Connexins/metabolism , Coronary Circulation , Heart/growth & development , Heart Diseases , Morphogenesis , Myocardium/cytology , Myocardium/metabolism , Purkinje Fibers/cytology , Signal Transduction/physiology , Gap Junction alpha-5 Protein
18.
Novartis Found Symp ; 250: 157-74; discussion 174-6, 276-9, 2003.
Article in English | MEDLINE | ID: mdl-12956329

ABSTRACT

We review here the evolution and development of the earliest components of the cardiac pacemaking and conduction system (PCS) and the turnover or persistence of such cells into old age in the adult vertebrate heart. Heart rate is paced by upstream foci of cardiac muscle near the future sinoatrial junction even before contraction begins. As the tubular heart loops, directional blood flow is maintained through coordinated sphincter function in the forming atrioventricular (AV) canal and outflow segments. Propagation of initially peristaltoid contraction along and between these segments appears to be influenced by physical conditioning and orientation of inner muscle layers as well as by their slow relaxation; all characteristic of definitive conduction tissue. As classical elements of the mature conduction system emerge, such inner 'contour fibres' maintain muscular and electrical continuity between atrial and ventricular compartments. Elements of such primordial architecture are visible also in histological and optical electrical study of fish and frog hearts. In the maturing chick heart, cells within core conducting tissues retain early thymidine labels from the tubular heart stage into adult life, dividing only slowly, if at all. Preliminary evidence from mammals suggest similar function and kinetics for these 'oldest, toughest' cells in the hearts of all vertebrates.


Subject(s)
Aging/physiology , Heart Conduction System/growth & development , Heart/anatomy & histology , Animals , Calcium/metabolism , Cell Differentiation/physiology , Heart/growth & development , Heart Conduction System/anatomy & histology , Heart Rate/physiology , Humans , Muscle Contraction/physiology , Myocardium/cytology , Myocardium/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism
19.
Anat Rec A Discov Mol Cell Evol Biol ; 274(1): 773-7, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12923887

ABSTRACT

Patterns of DNA synthesis in the developing mouse heart between ED7.5-18.5 were studied by a combination of thymidine and bromodeoxyuridine labeling techniques. From earliest stages, we found zones of slow myocyte proliferation at both the venous and arterial poles of the heart, as well as in the atrioventricular region. The labeling index was distinctly higher in nonmyocardial populations (endocardium, epicardium, and cardiac cushions). Ventricular trabeculae showed lower proliferative activity than the ventricular compact layer after their appearance at ED9.5. Low labeling was observed in the pectinate muscles of the atria from ED11.5. The His bundle, bundle branches, and Purkinje fiber network likewise were distinguished by their lack of labeling. Thymidine birthdating (label dilution) showed that the cells in these emerging components of the cardiac conduction system terminally differentiated between ED8.5-13.5. These patterns of slowed proliferation correlate well with those in other species, and can serve as a useful marker for the forming conduction system.


Subject(s)
Embryo, Mammalian/cytology , Heart Conduction System/embryology , Heart/embryology , Animals , Biomarkers/analysis , Cell Differentiation , Cell Division , Cellular Senescence , Embryo, Mammalian/physiology , Heart Conduction System/cytology , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Time Factors , Tissue Distribution
20.
Anat Rec A Discov Mol Cell Evol Biol ; 273(2): 687-99, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12845705

ABSTRACT

Marine mammals show many deviations from typical mammalian characteristics due to their high degree of specialization to the aquatic environment. In Cetaceans, some of the features of limbs and dentition resemble very ancestral patterns. In some species, hearts with a clearly bifid apex (a feature normally present during mammalian embryogenesis prior to completion of ventricular septation) have been described. However, there is a scant amount of data regarding heart development in Cetaceans, and it is not clear whether the bifid apex is the rule or the exception. We examined samples from a unique collection of embryonic dolphin specimens macroscopically and histologically to learn more about normal cardiac development in the spotted dolphin. It was found that during the dolphin's 280 days of gestation, the heart completes septation at about 35 days. However, substantial trabecular compaction, which normally occurs in chicks, mice, and humans at around that time period, was delayed until day 60, when coronary circulation became established. At that time, the apex still appeared bifid, similarly to early fetal mouse or rat hearts. By day 80, however, the heart gained a compacted, characteristic shape, with a single apex. It thus appears that the bifid apex in the adult Cetacean heart is probably particular to certain species, and its significance remains unclear.


Subject(s)
Body Patterning/physiology , Dolphins/embryology , Heart/embryology , Animals , Chick Embryo , Coronary Vessels/embryology , Coronary Vessels/physiology , Dolphins/physiology , Female , Heart/physiology , Heart Septum/embryology , Heart Ventricles/embryology , Humans , Mice , Myocardium/cytology , Rats , Ventricular Function
SELECTION OF CITATIONS
SEARCH DETAIL
...