Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
J Dev Biol ; 10(1)2022 Jan 10.
Article in English | MEDLINE | ID: mdl-35076524

ABSTRACT

Here, we have assembled five interesting manuscripts that deserve special attention [...].

2.
J Neurosci Res ; 100(12): 2127-2137, 2022 12.
Article in English | MEDLINE | ID: mdl-33687103

ABSTRACT

Developmental brain injury describes a spectrum of neurological pathologies resulting from either antenatal or perinatal injury. This includes both cognitive and motor defects that affect patients for their entire lives. Developmental brain injury can be caused by a spectrum of conditions including stroke, perinatal hypoxia-ischemia, and intracranial hemorrhage. Additional risk factors have been identified including very low birth weight, mechanical ventilation, and oxygen (O2 ) supplementation. In fact, infants with bronchopulmonary dysplasia, an inflammatory disease associated with disrupted lung development, have been shown to have decreased cerebral white matter and decreased intracranial volumes. Thus, there appears to be a developmental link between the lung, O2 , and the brain that leads to proper myelination. Here, we will discuss what is currently known about the link between O2 and myelination and how scientists are exploring mechanisms through which supplemental O2 and/or lung injury can affect brain development. Consideration of a link between the diseased lung and developing brain will allow clinicians to fine tune their approaches in managing preterm lung disease in order to optimize brain health.


Subject(s)
Brain Injuries , Lung Injury , White Matter , Infant, Newborn , Humans , Female , Pregnancy , White Matter/pathology , Oxygen , Lung Injury/complications , Lung Injury/pathology , Brain/pathology , Brain Injuries/pathology
3.
eNeuro ; 2021 Jun 07.
Article in English | MEDLINE | ID: mdl-34099489

ABSTRACT

Intrauterine growth restriction (IUGR) and oxygen exposure in isolation and combination adversely affect the developing brain, putting infants at risk for neurodevelopmental disability including cerebral palsy. Rodent models of IUGR and postnatal hyperoxia have demonstrated oligodendroglial injury with subsequent white matter injury (WMI) and motor dysfunction. Here we investigate transcriptomic dysregulation in IUGR with and without hyperoxia exposure to account for the abnormal brain structure and function previously documented. We performed RNA sequencing and analysis using a mouse model of IUGR and found that IUGR, hyperoxia, and the combination of IUGR with hyperoxia (IUGR/hyperoxia) produced distinct changes in gene expression. IUGR in isolation demonstrated the fewest differentially expressed genes compared to control. In contrast, we detected several gene alterations in IUGR/hyperoxia; genes involved in myelination were strikingly downregulated. We also identified changes to specific regulators including TCF7L2, BDNF, SOX2, and DGCR8, through Ingenuity Pathway Analysis, that may contribute to impaired myelination in IUGR/hyperoxia. Our findings show that IUGR with hyperoxia induces unique transcriptional changes in the developing brain. These indicate mechanisms for increased risk for WMI in IUGR infants exposed to oxygen and suggest potential therapeutic targets to improve motor outcomes.Significance StatementThis study demonstrates that perinatal exposures of IUGR and/or postnatal hyperoxia result in distinct transcriptomic changes in the developing brain. In particular, we found that genes involved in normal developmental myelination, myelin maintenance, and remyelination were most dysregulated when IUGR was combined with hyperoxia. Understanding how multiple risk factors lead to WMI is the first step in developing future therapeutic interventions. Additionally, because oxygen exposure is often unavoidable after birth, an understanding of gene perturbations in this setting will increase our awareness of the need for tight control of oxygen use to minimize future motor disability.

4.
Front Pediatr ; 7: 446, 2019.
Article in English | MEDLINE | ID: mdl-31781523

ABSTRACT

Fibrosis is an irreversible remodeling process characterized by the deposition of collagen in the extracellular matrix of various organs through a variety of pathologies in children, leading to the stiffening of healthy tissues and organ dysfunction. Despite the prevalence of fibrotic disease in children, large gaps exist in our understanding of the mechanisms that lead to fibrosis, and there are currently no therapies to treat or reverse it. We previously observed that castration significantly reduces fibrosis in the bladders of male mice that have been partially obstructed. Here, we investigated if the expression of androgen response genes were altered in mouse bladders after partial bladder outlet obstruction (PO). Using a QPCR microarray and QRTPCR we found that PO was sufficient to increase expression of the androgen response gene Nkx3.1. Consistent with this was an increase in the expression of NKX3.1 protein. Immunofluorescent antibody localization demonstrated nuclear NKX3.1 in most bladder cells after PO. We tested if genetic deletion of Nkx3.1 alters remodeling of the bladder wall after PO. After PO, Nkx3.1 KO/KO bladders underwent remodeling, demonstrating smaller bladder area, thickness, and bladder: body weight ratios than obstructed, wild type controls. Remarkably, Nkx3.1 KO/KO specifically affected histological parameters of fibrosis, including reduced collagen to muscle ratio. Loss of Nkx3.1 altered collagen and smooth muscle cytoskeletal gene expression following PO which supported our histologic findings. Together these findings indicated that after PO, Nkx3.1 expression is induced in the bladder and that it mediates important pathways that lead to tissue fibrosis. As Nkx3.1 is an androgen response gene, our data suggest a possible mechanism by which fibrosis is mediated in male mice and opens the possibility of a molecular pathway mediated by NKX3.1 that could explain sexual dimorphism in bladder fibrosis.

5.
Am J Physiol Renal Physiol ; 317(6): F1503-F1512, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31532245

ABSTRACT

We have defined a population of stem cell antigen (Sca)-1+/CD34+/lin- mesenchymal stem cells in the mouse urinary bladder. These cells are reduced after partial bladder outlet obstruction (PO). To test the role of Sca-1 expressed by these cells, we analyzed bladders from Sca-1 knockout (KO) mice in both uninjured male mice and male mice subjected to PO. We found that loss of Sca-1 alone had little effect on bladder development or function but reduced the total number of mesenchymal stem cells by 30%. After PO, bladders from Sca-1-null KO male mice were larger, with more collagen and less muscle, than obstructed wild-type mice. Steady-state levels of caldesmon were significantly reduced and levels of fibroblast-specific protein 1 were significantly increased in Sca-1 KO mice compared with wild-type mice after PO. In investigating the effects of PO on cell proliferation, we found that loss of Sca-1 changed the timing of cell division in CD34+/lin-, collagen-producing, and smooth muscle cells. PO in combination with loss of Sca-1 drastically reduced the ability of CD34+/lin- cells to form colonies in vitro. Our findings therefore support the hypothesis that Sca-1 protects the bladder from fibrotic remodeling after obstruction, in part by influencing the proliferation of cells responding to the injury.


Subject(s)
Antigens, Ly/therapeutic use , Membrane Proteins/therapeutic use , Urinary Bladder/pathology , Animals , Antigens/immunology , Antigens/therapeutic use , Antigens, CD34/metabolism , Antigens, Ly/genetics , Antigens, Ly/immunology , Calmodulin-Binding Proteins/metabolism , Cell Proliferation , Fibrosis , Male , Membrane Proteins/genetics , Membrane Proteins/immunology , Mesenchymal Stem Cells/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/metabolism , Protective Agents , Stem Cells , Urinary Bladder Neck Obstruction/pathology
6.
Am J Clin Exp Urol ; 6(5): 189-196, 2018.
Article in English | MEDLINE | ID: mdl-30510971

ABSTRACT

The partial bladder outlet obstruction animal model (pBOO) is commonly used as a model for obstructive uropathy. Unfortunately, pBOO demonstrates variable degrees of obstruction requiring bladder weight (BW) or urodynamic studies to determine true obstruction. Our objective is to identify extent of obstruction by correlating early post-operative Void Stains on Paper (VSOP) assays with ultimate BW in mice. pBOO was performed on 32 mice 1- and 4-week VSOPs were quantified for mean voided volume (mVV). At 4 weeks, bladders were harvested and weighed. Correlation was evaluated through bivariate kernel density estimation and a Pearson correlation coefficient (SAS). Single variable histogram of the data established groups based on BWs and mVV. mVV's and bladder weights within group pairings were averaged and plotted to render a non-linear regression model. A significant correlation was found between 1-week mVVs and 4-week BWs upon bivariate analysis with a correlation coefficient of -0.758 (p = 0.0294). A non-linear regression of plotted data defined a statistically significant fit equation correlating 1-week mVV to 4-week BW. We demonstrate a novel method for forecasting degree of obstruction in pBOO based on 1-week post-operative VSOP mVV.

7.
PLoS One ; 13(11): e0206436, 2018.
Article in English | MEDLINE | ID: mdl-30475828

ABSTRACT

Cystectomy is the removal of all or part of the urinary bladder. It has been observed that there is significant regrowth of the bladder after partial cystectomy and this has been proposed to be through regeneration of the organ. Regrowth of tissue in mammals has been proposed to involve compensatory mechanisms that share many characteristics of true regeneration, like the growth of specialized structures such as blood vessels or nerves. However, the overall structure of the normal organ is not achieved. Here we tested if bladder growth after subtotal cystectomy (STC, removal of 50% of the bladder) was compensatory or regenerative. To do this we subjected adult female mouse bladders to STC and assessed regrowth using several established cellular parameters including histological, gene expression, cytokine accumulation and cell proliferation studies. Bladder function was analyzed using cystometry and the voiding stain on paper (VSOP) technique. We found that STC bladders were able to increase their ability to hold urine with the majority of volume restoration occurring within the first two weeks. Regenerating bladders had thinner walls with less mean muscle thickness, and they showed increased collagen deposition at the incision as well as throughout the bladder wall suggesting that fibrosis was occurring. Cell populations differed in their response to injury with urothelial regeneration complete by day 7, but stromal and detrusor muscle still incomplete after 8wks. Cells incorporated EdU when administered at the time of surgery and tracing of EdU positive cells over time indicated that many newborn cells originate at the incision and move mediolaterally. Basal urothelial cells and bladder mesenchymal stem cells but not smooth muscle cells significantly incorporated EdU after STC. Since anti-inflammatory cytokines play a role in regeneration, we analyzed expressed cytokines and found that no anti-inflammatory cytokines were present in the bladder 1wk after STC. Our findings suggest that bladder regrowth after cystectomy is compensatory and functions to increase the volume that the bladder can hold. This finding sets the stage for understanding how the bladder responds to cystectomy and how this can be improved in patients after suffering bladder injury.


Subject(s)
Cystectomy , Regeneration , Urinary Bladder/physiology , Urinary Bladder/surgery , Animals , Cicatrix/etiology , Cicatrix/genetics , Collagen/metabolism , Cystectomy/adverse effects , Cytokines/metabolism , Female , Fibrosis , Gene Expression Regulation , Mice , Recovery of Function , Urinary Bladder/metabolism , Urinary Bladder/pathology
8.
Dev Neurosci ; 40(4): 344-357, 2018.
Article in English | MEDLINE | ID: mdl-30428455

ABSTRACT

Intrauterine growth restriction (IUGR) is estimated to occur in 5% of pregnancies, with placental insufficiency being the most common cause in developed countries. While it is known that white matter injury occurs in premature infants, the extent of IUGR on white matter injury is less defined in term infants. We used a novel murine model that utilizes a thromboxane A2 (TXA2) analog (U46619), a potent vasoconstrictor, to induce maternal hypertension and mimic human placental insufficiency-induced IUGR to study the white matter. We also investigated the role of hyperoxia as an additional risk factor for white matter injury, as IUGR infants are at increased risk of respiratory comorbidities leading to increased oxygen exposure. We found that TXA2 analog-induced IUGR results in white matter injury as demonstrated by altered myelin structure and changes in the oligodendroglial cell/oligodendrocyte population. In addition, our study demonstrates that hyperoxia exposure independently results in white matter perturbation. To our knowledge, this is the first study to report single and combined effects of IUGR with hyperoxia impacting the white matter and motor function. These results draw attention to the need for close monitoring of motor development in IUGR babies following hospital discharge as well as highlighting the importance of limiting, as clinically feasible, the degree of oxygen overexposure to potentially improve motor outcomes in this population of infants.


Subject(s)
Brain/growth & development , Fetal Growth Retardation/physiopathology , Hyperoxia/metabolism , Infant, Premature/growth & development , White Matter/injuries , Animals , Animals, Newborn , Brain Injuries/etiology , Female , Mice, Inbred C57BL , Placental Insufficiency/metabolism , Pregnancy , White Matter/physiopathology
10.
Dev Neurosci ; 40(1): 23-38, 2018.
Article in English | MEDLINE | ID: mdl-29324456

ABSTRACT

Hypoxic-ischemic injury (HI) to the neonatal human brain results in myelin loss that, in some children, can manifest as cerebral palsy. Previously, we had found that neuronal overexpression of the bone morphogenic protein (BMP) inhibitor noggin during development increased oligodendroglia and improved motor function in an experimental model of HI utilizing unilateral common carotid artery ligation followed by hypoxia. As BMPs are known to negatively regulate oligodendroglial fate specification of neural stem cells and alter differentiation of committed oligodendroglia, BMP signaling is likely an important mechanism leading to myelin loss. Here, we showed that BMP signaling is upregulated within oligodendroglia of the neonatal brain. We tested the hypothesis that inhibition of BMP signaling specifically within neural progenitor cells (NPCs) is sufficient to protect oligodendroglia. We conditionally deleted the BMP receptor 2 subtype (BMPR2) in NG2-expressing cells after HI. We found that BMPR2 deletion globally protects the brain as assessed by MRI and protects motor function as assessed by digital gait analysis, and that conditional deletion of BMPR2 maintains oligodendrocyte marker expression by immunofluorescence and Western blot and prevents loss of oligodendroglia. Finally, BMPR2 deletion after HI results in an increase in noncompacted myelin. Thus, our data indicate that inhibition of BMP signaling specifically in NPCs may be a tractable strategy to protect the newborn brain from HI.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Brain/metabolism , Hypoxia-Ischemia, Brain/metabolism , Motor Activity/physiology , Neural Stem Cells/metabolism , Animals , Animals, Newborn , Gene Knockdown Techniques , Hypoxia-Ischemia, Brain/pathology , Mice , Mice, Inbred C57BL , Oligodendroglia/metabolism , Signal Transduction/physiology
11.
Dev Dyn ; 247(3): 332-339, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28786157

ABSTRACT

The collagen gel has been used to study epithelial-mesenchymal transformation (EMT) for over 30 years. With advances in the field of materials sciences, new options are available to design optically clear, three-dimensional nature-inspired matrix mimetics to study EMT. Here, we review the history of the collagen gel assay, discuss its current use and how newer artificial matrices can be built to simulate in vivo extracellular environments and investigate important current questions in the EMT field. We suggest that further collaborations between materials scientists and biologists will be critical to move the field of EMT forward. Developmental Dynamics 247:332-339, 2018. © 2017 Wiley Periodicals, Inc.


Subject(s)
Biological Assay/history , Epithelial-Mesenchymal Transition , Hydrogels/chemistry , Biological Assay/methods , Collagen , History, 20th Century , History, 21st Century , Humans , Methods
12.
Front Pediatr ; 5: 132, 2017.
Article in English | MEDLINE | ID: mdl-28638819

ABSTRACT

Lower urinary tract symptoms secondary to posterior urethral valves (PUV) arise in boys during adolescence. The reasons for this have previously been attributed to increased urine output as boys experience increased growth. Additionally, there are few choices for clinicians to effectively treat these complications. We formed the new hypothesis that increased androgen levels at this time of childhood development could play a role at the cellular level in obstructed bladders. To test this hypothesis, we investigated the role of testosterone on bladder detrusor muscle following injury from partial bladder outlet obstruction (PO) in mice. A PO model was surgically created in juvenile male mice. A group of mice were castrated by bilateral orchiectomy at time of obstruction (CPO). Testosterone cypionate was administered to a group of castrated, obstructed mice (CPOT). Bladder function was assessed by voiding stain on paper (VSOP). Bladders were analyzed at 7 and 28 days by weight and histology. Detrusor collagen to smooth muscle ratio (Col/SM) was calculated using Masson's trichrome stain. All obstructed groups had lower max voided volumes (MVV) than sham mice at 1 day. Hormonally intact mice (PO) continued to have lower MVV at 7 and 28 days while CPO mice improved to sham levels at both time points. In accordance, PO mice had higher bladder-to-body weight ratios than CPO and sham mice demonstrating greater bladder hypertrophy. Histologically, Col/SM was lower in sham and CPO mice. When testosterone was restored in CPOT mice, MVV remained low at 7 and 28 days compared to CPO and bladder-to-body weight ratios were also greater than CPO. Histologic changes were also seen in CPOT mice with higher Col/SM than sham and CPO mice. In conclusion, our findings support a role for testosterone in the fibrotic changes that occur after obstruction in male mice. This suggests that while other changes may occur in adolescent boys that cause complication in boys with PUV, the bladder itself responds to testosterone at the cellular level. This opens the door to a new understanding of pathways that influence bladder fibrosis and could lead to novel approaches to treat boys with PUV.

13.
PLoS One ; 10(11): e0141437, 2015.
Article in English | MEDLINE | ID: mdl-26540309

ABSTRACT

Bladder fibrosis is an undesired end point of injury of obstruction and often renders the smooth muscle layer noncompliant. In many cases, the long-term effect of bladder fibrosis is renal failure. Despite our understanding of the progression of this disease, little is known about the cellular mechanisms that lead to a remodeled bladder wall. Resident stem (progenitor) cells have been identified in various organs such as the brain, heart and lung. These cells function normally during organ homeostasis, but become dysregulated after organ injury. Here, we aimed to characterize a mesenchymal progenitor cell population as a first step in understanding its role in bladder fibrosis. Using fluorescence activated cell sorting (FACS), we identified a Sca-1+/ CD34+/ lin- (PECAM-: CD45-: Ter119-) population in the adult murine bladder. These cells were localized to the stromal layer of the adult bladder and appeared by postnatal day 1. Cultured Sca-1+/ CD34+/ lin- bladder cells self-renewed, formed colonies and spontaneously differentiated into cells expressing smooth muscle genes. These cells differentiated into other mesenchymal lineages (chondrocytes, adipocytes and osteocytes) upon culture in induction medium. Both acute and partial obstruction of the bladder reduced expression of CD34 and changed localization of Sca-1 to the urothelium. Partial obstruction resulted in upregulation of fibrosis genes within the Sca-1+/CD34+/lin- population. Our data indicate a resident, mesenchymal stem cell population in the bladder that is altered by bladder obstruction. These findings provide new information about the cellular changes in the bladder that may be associated with bladder fibrosis.


Subject(s)
Mesenchymal Stem Cells/physiology , Urinary Bladder/cytology , Animals , Antigens, CD34/metabolism , Antigens, Ly/metabolism , Cell Lineage , Cells, Cultured , Fibrosis , Flow Cytometry , Fluorescent Antibody Technique , Membrane Proteins/metabolism , Mice , Polymerase Chain Reaction , Urinary Bladder/pathology , Urinary Bladder Neck Obstruction/pathology
14.
Am J Respir Crit Care Med ; 189(3): 314-24, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24251580

ABSTRACT

RATIONALE: Chronic hypoxia induces pulmonary vascular remodeling, pulmonary hypertension, and right ventricular hypertrophy. At present, little is known about mechanisms driving these responses. Hypoxia-inducible factor-1α (HIF-1α) is a master regulator of transcription in hypoxic cells, up-regulating genes involved in energy metabolism, proliferation, and extracellular matrix reorganization. Systemic loss of a single HIF-1α allele has been shown to attenuate hypoxic pulmonary hypertension, but the cells contributing to this response have not been identified. OBJECTIVES: We sought to determine the contribution of HIF-1α in smooth muscle on pulmonary vascular and right heart responses to chronic hypoxia. METHODS: We used mice with homozygous conditional deletion of HIF-1α combined with tamoxifen-inducible smooth muscle-specific Cre recombinase expression. Mice received either tamoxifen or vehicle followed by exposure to either normoxia or chronic hypoxia (10% O2) for 30 days before measurement of cardiopulmonary responses. MEASUREMENTS AND MAIN RESULTS: Tamoxifen-induced smooth muscle-specific deletion of HIF-1α attenuated pulmonary vascular remodeling and pulmonary hypertension in chronic hypoxia. However, right ventricular hypertrophy was unchanged despite attenuated pulmonary pressures. CONCLUSIONS: These results indicate that HIF-1α in smooth muscle contributes to pulmonary vascular remodeling and pulmonary hypertension in chronic hypoxia. However, loss of HIF-1 function in smooth muscle does not affect hypoxic cardiac remodeling, suggesting that the cardiac hypertrophy response is not directly coupled to the increase in pulmonary artery pressure.


Subject(s)
Hypertension, Pulmonary/metabolism , Hypertrophy, Right Ventricular/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/complications , Muscle, Smooth, Vascular/metabolism , Pulmonary Artery/metabolism , Airway Remodeling , Animals , Chronic Disease , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/pathology , Hypertrophy, Right Ventricular/etiology , Hypertrophy, Right Ventricular/pathology , Hypoxia/metabolism , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Male , Mice , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Pulmonary Artery/pathology , Random Allocation
15.
Dev Biol ; 366(2): 111-24, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22546693

ABSTRACT

The importance of the epicardium for myocardial and valvuloseptal development has been well established; perturbation of epicardial development results in cardiac abnormalities, including thinning of the ventricular myocardial wall and malformations of the atrioventricular valvuloseptal complex. To determine the spatiotemporal contribution of epicardially derived cells to the developing fibroblast population in the heart, we have used a mWt1/IRES/GFP-Cre mouse to trace the fate of EPDCs from embryonic day (ED)10 until birth. EPDCs begin to populate the compact ventricular myocardium around ED12. The migration of epicardially derived fibroblasts toward the interface between compact and trabecular myocardium is completed around ED14. Remarkably, epicardially derived fibroblasts do not migrate into the trabecular myocardium until after ED17. Migration of EPDCs into the atrioventricular cushion mesenchyme commences around ED12. As development progresses, the number of EPDCs increases significantly, specifically in the leaflets which derive from the lateral atrioventricular cushions. In these developing leaflets the epicardially derived fibroblasts eventually largely replace the endocardially derived cells. Importantly, the contribution of EPDCs to the leaflets derived from the major AV cushions is very limited. The differential contribution of EPDCs to the various leaflets of the atrioventricular valves provides a new paradigm in valve development and could lead to new insights into the pathogenesis of abnormalities that preferentially affect individual components of this region of the heart. The notion that there is a significant difference in the contribution of epicardially and endocardially derived cells to the individual leaflets of the atrioventricular valves has also important pragmatic consequences for the use of endocardial and epicardial cre-mouse models in studies of heart development.


Subject(s)
Fibroblasts/cytology , Heart Valves/embryology , Heart/embryology , Pericardium/cytology , Animals , Embryonic Development , Heart Valves/cytology , Heart Ventricles/cytology , Heart Ventricles/embryology , Mice , Organogenesis
16.
J Cell Sci ; 123(Pt 3): 431-40, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20067998

ABSTRACT

The transient and localized signaling events between invasive breast cancer cells and the underlying endothelial cells have remained poorly characterized. We report a novel approach integrating vascular engineering with three-dimensional time-lapse fluorescence resonance energy transfer (FRET) imaging to dissect how endothelial myosin light chain kinase (MLCK) is modulated during tumor intravasation. We show that tumor transendothelial migration occurs via both paracellular (i.e. through cell-cell junctions) and transcellular (i.e. through individual endothelial cells) routes. Endothelial MLCK is activated at the invasion site, leading to regional diphosphorylation of myosin-II regulatory light chain (RLC) and myosin contraction. Blocking endothelial RLC diphosphorylation blunts tumor transcellular, but not paracellular, invasion. Our results implicate an important role for endothelial myosin-II function in tumor intravasation.


Subject(s)
Breast Neoplasms/enzymology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Fluorescence Resonance Energy Transfer/methods , Imaging, Three-Dimensional/methods , Myosin-Light-Chain Kinase/metabolism , Neoplasm Invasiveness/pathology , Animals , Breast Neoplasms/pathology , Cattle , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/physiology , Humans , Microscopy, Confocal , Myosin-Light-Chain Kinase/genetics , Phosphorylation
17.
Dev Dyn ; 239(2): 598-609, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19918883

ABSTRACT

Little is known about the molecules that mediate the attachment of proepicardial cells to the heart. Ephrins are cell surface ligands for Eph tyrosine kinase receptors, molecules known to play a role in cell adhesion and migration. Here, we detected EphrinB ligands in proepicardial and epicardial mesothelial cells (EMCs) using reverse transcriptase-polymerase chain reaction, immunoblotting, immunolocalization, and EphB-Fc binding. Aggregated EphB-Fc fragments clustered ephrinB1 ligands on living EMCs indicating that they are cell surface expressed. In vitro assays demonstrated that ephrinB ligands participate in EMC migration but not cell adhesion. Localization studies in hearts at Hamburger and Hamilton stage 30 and older revealed that ephrinB1 is expressed in the epicardium and subepicardial mesenchyme of the atrioventricular sulcus. EMCs treated with platelet-derived growth factor-BB expressed smooth muscle markers but not ephrinB1. Our study supports an early role for ephrinB ligands for migration of epicardial cells and a later role in perivascular fibroblasts of coronary vessels in the atrioventricular sulcus.


Subject(s)
Cell Movement , Coronary Vessels/embryology , Ephrin-B1/metabolism , Ephrin-B2/metabolism , Pericardium/embryology , Animals , Cell Adhesion , Chick Embryo , Coronary Vessels/metabolism , Fibroblasts/metabolism , Mice , Myocardium/metabolism , Pericardium/metabolism , Rats , Receptors, Eph Family/metabolism
18.
Dev Dyn ; 238(2): 423-30, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19161222

ABSTRACT

Though development of the coronary vasculature is a critical event during embryogenesis, the molecular mechanisms that regulate its formation are not well characterized. Two unique approaches were used to investigate interactions between cardiac myocytes and proepicardial (PE) cells, which are the coronary anlagen. One of these experimental approaches used a 3-D collagen scaffold system on which specific cell-cell and cell-matrix interactions were studied. The other approach used a whole heart culture system that allowed for the analysis of epicardial to mesenchymal transformation (EMT). The VEGF signaling system has been implicated previously as an important regulator of coronary development. Our results demonstrated that a specific isoform of VEGF-A, VEGF(164), increased PE-derived endothelial cell proliferation and also increased EMT. However, VEGF-stimulated endothelial cells did not robustly coalesce into endothelial tubes as they did when cocultured with cardiac myocytes. Interestingly, blocking VEGF signaling via flk-1 inhibition reduced endothelial tube formation despite the presence of cardiac myocytes. These results indicate that VEGF signaling is complex during coronary development and that combinatorial signaling by other VEGF-A isoforms or other flk-1-binding VEGFs are likely to regulate endothelial tube formation.


Subject(s)
Coronary Vessels/physiology , Endothelium, Vascular/physiology , Myocytes, Cardiac/physiology , Vascular Endothelial Growth Factor A/physiology , Animals , Cell Proliferation , Cells, Cultured , Chickens , Collagen , Coronary Vessels/cytology , Endothelium, Vascular/cytology , Mice , Morphogenesis/physiology , Myocytes, Cardiac/metabolism , Organ Culture Techniques , Pericardium/cytology , Pericardium/physiology , Protein Isoforms/metabolism , Quail , Vascular Endothelial Growth Factor Receptor-2/metabolism
19.
Dev Dyn ; 237(4): 962-78, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18351655

ABSTRACT

Formation of the epicardium requires interactions between alpha(4)beta(1) integrin, and the extracellular matrix. We investigated the role of other integrins expressed by epicardial cells. We detected transcripts for alpha(5), alpha(8), alpha(v), beta(1), beta(3), and beta(5) integrins in the chick proepicardial organ (PE). We demonstrate that alpha(5)beta(1), alpha(8)beta(1), and alpha(v)beta(3) integrins are expressed by chick epicardial mesothelial cells (EMCs). Migration of EMCs in vitro was reduced by RGD-containing peptides. Using adenoviruses expressing an antisense to chick alpha(4) (AdGFPalpha4AS), full-length (Adhalpha4V5), and C-terminal deleted alpha(4) (Adhalpha4DeltaCV5), we found that EMCs were less able to adhere to vitronectin and fibronectin(120) indicating that alpha(4)beta(1) plays a role in regulating EMC adhesion to ligands of alpha(5)beta(1), alpha(8)beta(1), and alpha(v)beta(3). In Adhalpha4DeltaCV5-infected EMCs, alpha(5)beta(1) was diminished in fibrillar adhesions and new FN matrix assembly was abnormal. We propose that cooperation between alpha(4)beta(1) and RGD integrins is important for EMC adhesion and subepicardial matrix formation.


Subject(s)
Cell Adhesion/physiology , Extracellular Matrix/metabolism , Integrin alpha4beta1/metabolism , Integrin alpha5beta1/metabolism , Pericardium/cytology , Pericardium/embryology , Animals , Cell Movement/physiology , Chick Embryo/anatomy & histology , Chick Embryo/metabolism , Coronary Vessels/anatomy & histology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Epithelium/anatomy & histology , Epithelium/metabolism , Extracellular Matrix/chemistry , Fibronectins/metabolism , Humans , Integrin alpha4beta1/genetics , Integrin alpha5beta1/genetics , Oligopeptides/metabolism , Pericardium/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Vitronectin/metabolism
20.
Dev Biol ; 302(1): 230-42, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17045582

ABSTRACT

The T-box transcription factor Tbx5 can interact with Nkx2.5 and Gata4 transcription factors to synergistically regulate heart-specific genes in the nucleus. While a nuclear role for Tbx5 is clearly defined, we have previously shown that Tbx5 shuttles from nuclear to cytoplasmic sites, forming a complex with the PDZ-LIM protein LMP4 on the actin cytoskeleton. In this study, using a developmental series of chicken hearts, we provide the first evidence for differential Tbx5 protein expression and sub-cellular localization during cardiogenesis. At the tissue level, we show temporally and spatially restricted Tbx5 co-expression with LMP4. In cells co-expressing LMP4 and Tbx5 we demonstrate dynamic Tbx5 re-localization from exclusively nuclear to nuclear and cytoplasmic expression in the atrio-ventricular cushion. Furthermore, in coronary vessel development we show exclusive cytoplasmic localization of Tbx5, indicating a function for Tbx5 in the cytoplasm. In addition, we discover unknown regulation of Tbx5 and LMP4 expression in epicardial tissue, suggesting a specific role for Tbx5 in epicardial formation. These studies provide in vivo significance of the LMP4/Tbx5 protein interaction, suggesting both nuclear and cytoplasmic roles for Tbx5. The shuttling between nuclear and cytoplasmic sites reveals a novel mechanism for Tbx transcription factor regulation in chicken heart development allowing new insights for a better understanding of the molecular basis of hand/heart birth defects associated with TBX5 mutations.


Subject(s)
Gene Expression Regulation, Developmental , Heart/embryology , Myocardium/metabolism , T-Box Domain Proteins/genetics , Active Transport, Cell Nucleus , Animals , Cell Nucleus/metabolism , Chick Embryo , Coronary Vessels/embryology , Myocardium/chemistry , Myocardium/cytology , Pericardium/embryology , Protein Binding , Protein Transport , Proteins/genetics , Proteins/metabolism , T-Box Domain Proteins/analysis , T-Box Domain Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...