1
|
Lungu CN, Mehedinti MC. Molecular Motifs in Vascular Morphogenesis: Vascular Endothelial Growth Factor A (VEGFA) as the Leading Promoter of Angiogenesis. Int J Mol Sci 2023; 24:12169. [PMID: 37569543 PMCID: PMC10418718 DOI: 10.3390/ijms241512169] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Tissular hypoxia stimulates vascular morphogenesis. Vascular morphogenesis shapes the cell and, consecutively, tissue growth. The development of new blood vessels is intermediated substantially through the tyrosine kinase pathway. There are several types of receptors inferred to be located in the blood vessel structures. Vascular endothelial growth factor A (VEGF-A) is the leading protagonist of angiogenesis. VEGF-A's interactions with its receptors VEGFR1, VEGFR2, and VEGFR3, together with disintegrin and metalloproteinase with thrombospondin motifs 1 (ADAMTS1), connective tissue growth factor (CTGF), and neuropilin-1 (NRP1), independently, are studied computationally. Peripheral artery disease (PAD), which results in tissue ischemia, is more prevalent in the senior population. Presently, medical curatives used to treat cases of PAD-antiplatelet and antithrombotic agents, statins, antihypertensive remedies with ACE (angiotensin-converting enzyme) impediments, angiotensin receptor blockers (ARB) or β- blockers, blood glucose control, and smoking cessation-are not effective. These curatives were largely established from the treatment of complaint cases of coronary disease. However, these medical curatives do not ameliorate lower limb perfusion in cases of PAD. Likewise, surgical or endovascular procedures may be ineffective in relieving symptoms. Eventually, after successful large vessel revascularization, the residual microvascular circulation may well limit the effectiveness of curatives in cases of PAD. It would thus feel rational to attempt to ameliorate perfusion in PAD by enhancing vascular rejuvenescence and function. Likewise, stimulating specific angiogenesis in these cases (PAD) can ameliorate the patient's symptomatology. Also, the quality of life of PAD patients can be improved by developing new vasodilative and angiogenetic molecules that stimulate the tyrosine kinase pathway. In this respect, the VEGFA angiogenetic pathway was explored computationally. Docking methodologies, molecular dynamics, and computational molecular design methodologies were used. VEGFA's interaction with its target was primarily studied. Common motifs in the vascular morphogenesis pathway are suggested using conformational energy and Riemann spaces. The results show that interaction with VEGFR2 and ADAMTS1 is pivotal in the angiogenetic process. Also, the informational content of two VEGFA complexes, VEGFR2 and ADAMTS1, is crucial in the angiogenesis process.
Collapse
Affiliation(s)
- Claudiu N. Lungu
- Departament of Functional and Morphological Science, Faculty of Medicine and Pharamacy, Dunarea de Jos University, 800010 Galati, Romania
| | | |
Collapse
|
2
|
VanHeyst KA, Choi SH, Kingsley DT, Huang AY. Ectopic Tumor VCAM-1 Expression in Cancer Metastasis and Therapy Resistance. Cells 2022; 11:cells11233922. [PMID: 36497180 PMCID: PMC9735769 DOI: 10.3390/cells11233922] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 11/23/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Vascular Cell Adhesion Molecule-1 (VCAM-1; CD106) is a membrane protein that contributes critical physiologic functional roles in cellular immune response, including leukocyte extravasation in inflamed and infected tissues. Expressed as a cell membrane protein, VCAM-1 can also be cleaved from the cell surface into a soluble form (sVCAM-1). The integrin α4β1 (VLA-4) was identified as the first major ligand for VCAM-1. Ongoing studies suggest that, in addition to mediating physiologic immune functions, VCAM-1/VLA-4 signaling plays an increasingly vital role in the metastatic progression of various tumors. Additionally, elevated concentrations of sVCAM-1 have been found in the peripheral blood of patients with cancer, suggesting the tumor microenvironment (TME) as the source of sVCAM-1. Furthermore, over-expression of VLA-4 was linked to tumor progression in various malignancies when VCAM-1 was also up-regulated. This review explores the functional role of VCAM-1 expression in cancer metastasis and therapy resistance, and the potential for the disruption of VCAM-1/VLA-4 signaling as a novel immunotherapeutic approach in cancer, including osteosarcoma, which disproportionately affects the pediatric, adolescent and young adult population, as an unmet medical need.
Collapse
Affiliation(s)
- Kristen A. VanHeyst
- Center for Pediatric Immunotherapy at Rainbow, Angie Fowler AYA Cancer Institute, Division of Pediatric Hematology-Oncology, UH Rainbow Babies and Children’s Hospital, Cleveland, OH 44106, USA
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Sung Hee Choi
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | - Alex Y. Huang
- Center for Pediatric Immunotherapy at Rainbow, Angie Fowler AYA Cancer Institute, Division of Pediatric Hematology-Oncology, UH Rainbow Babies and Children’s Hospital, Cleveland, OH 44106, USA
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Correspondence: ; Tel.: +1-216-368-1271
| |
Collapse
|
3
|
Faralli JA, Filla MS, Peters DM. Integrin Crosstalk and Its Effect on the Biological Functions of the Trabecular Meshwork/Schlemm’s Canal. Front Cell Dev Biol 2022; 10:886702. [PMID: 35573686 PMCID: PMC9099149 DOI: 10.3389/fcell.2022.886702] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/05/2022] [Indexed: 11/20/2022] Open
Abstract
Integrins are a family of heterodimeric receptors composed of an α- and β-subunit that mediate cell-adhesion to a number of extracellular matrix (ECM) proteins in the Trabecular Meshwork/Schlemm’s canal (TM/SC) of the eye. Upon binding an ECM ligand, integrins transmit signals that activate a number of signaling pathways responsible for regulating actin-mediated processes (i.e phagocytosis, cell contractility, and fibronectin fibrillogenesis) that play an important role in regulating intraocular pressure (IOP) and may be involved in glaucoma. An important function of integrin-mediated signaling events is that the activity of one integrin can affect the activity of other integrins in the same cell. This creates a crosstalk that allows TM/SC cells to respond to changes in the ECM presumably induced by the mechanical forces on the TM/SC, aging and disease. In this review, we discuss how integrin crosstalk influences the function of the human TM/SC pathway. In particular, we will discuss how different crosstalk pathways mediated by either the αvβ3 or α4β1 integrins can play opposing roles in the TM when active and therefore act as on/off switches to modulate the cytoskeleton-mediated processes that regulate the outflow of aqueous humor through the TM/SC.
Collapse
Affiliation(s)
- Jennifer A. Faralli
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Mark S. Filla
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Donna M. Peters
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- *Correspondence: Donna M. Peters,
| |
Collapse
|
4
|
Affiliation(s)
- Laura Airas
- Division of Clinical Neurosciences, Turku University Hospital, Turku, Finland; Department of Clinical Medicine, University of Turku, Turku, Finland/Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| |
Collapse
|
5
|
Heparanase-induced shedding of syndecan-1/CD138 in myeloma and endothelial cells activates VEGFR2 and an invasive phenotype: prevention by novel synstatins. Oncogenesis 2016; 5:e202. [PMID: 26926788 PMCID: PMC5154350 DOI: 10.1038/oncsis.2016.5] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 12/15/2016] [Accepted: 01/13/2016] [Indexed: 12/17/2022] Open
Abstract
Multiple myeloma arises when malignant plasma cells invade and form multiple tumors in the bone marrow. High levels of heparanase (HPSE) correlate with poor prognosis in myeloma patients. A likely target of the enzyme is the heparan sulfate (HS) proteoglycan syndecan-1 (Sdc1, CD138), which is highly expressed on myeloma cells and contributes to poor prognosis in this disease. We find that HPSE promotes an invasive phenotype mediated by the very late antigen-4 (VLA-4, or α4β1 integrin) in myeloma cells plated on either fibronectin (FN) or vascular endothelial cell adhesion molecule-1 (VCAM-1), ligands that are prevalent in the bone marrow. The phenotype depends on vascular endothelial cell growth factor receptor-2 (VEGFR2), which is aberrantly expressed in myeloma, and is characterized by a highly protrusive lamellipodium and cell invasion. HPSE-mediated trimming of the HS on Sdc1 and subsequent matrix metalloproteinase-9-mediated shedding of the syndecan exposes a juxtamembrane site in Sdc1 that binds VEGFR2 and VLA-4, thereby coupling VEGFR2 to the integrin. Shed Sdc1 can be mimicked by recombinant Sdc1 ectodomain or by a peptide based on its binding motif, which causes VLA-4 to re-orient from the lagging edge (uropod) to the leading edge of migrating cells, couple with and activate VEGFR2. Peptides (called 'synstatins') containing only the VLA-4 or VEGFR2 binding sites competitively inhibit invasion, as they block coupling of the receptors. This mechanism is also utilized by vascular endothelial cells, in which it is also activated by HPSE, during endothelial cell tube formation. Collectively, our findings reveal for the first time the mechanism through which HPSE modulates Sdc1 function to promote both tumor cell invasion and angiogenesis, thereby driving multiple myeloma progression. The inhibitory synstatins, or inhibitors of HPSE enzyme activity, are likely to show promise as therapeutics against myeloma extravasation and spread.
Collapse
|
6
|
Vecino E, Rodriguez FD, Ruzafa N, Pereiro X, Sharma SC. Glia-neuron interactions in the mammalian retina. Prog Retin Eye Res 2015; 51:1-40. [PMID: 26113209 DOI: 10.1016/j.preteyeres.2015.06.003] [Citation(s) in RCA: 538] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 05/18/2015] [Accepted: 06/02/2015] [Indexed: 02/07/2023]
Abstract
The mammalian retina provides an excellent opportunity to study glia-neuron interactions and the interactions of glia with blood vessels. Three main types of glial cells are found in the mammalian retina that serve to maintain retinal homeostasis: astrocytes, Müller cells and resident microglia. Müller cells, astrocytes and microglia not only provide structural support but they are also involved in metabolism, the phagocytosis of neuronal debris, the release of certain transmitters and trophic factors and K(+) uptake. Astrocytes are mostly located in the nerve fibre layer and they accompany the blood vessels in the inner nuclear layer. Indeed, like Müller cells, astrocytic processes cover the blood vessels forming the retinal blood barrier and they fulfil a significant role in ion homeostasis. Among other activities, microglia can be stimulated to fulfil a macrophage function, as well as to interact with other glial cells and neurons by secreting growth factors. This review summarizes the main functional relationships between retinal glial cells and neurons, presenting a general picture of the retina recently modified based on experimental observations. The preferential involvement of the distinct glia cells in terms of the activity in the retina is discussed, for example, while Müller cells may serve as progenitors of retinal neurons, astrocytes and microglia are responsible for synaptic pruning. Since different types of glia participate together in certain activities in the retina, it is imperative to explore the order of redundancy and to explore the heterogeneity among these cells. Recent studies revealed the association of glia cell heterogeneity with specific functions. Finally, the neuroprotective effects of glia on photoreceptors and ganglion cells under normal and adverse conditions will also be explored.
Collapse
Affiliation(s)
- Elena Vecino
- Department of Cell Biology and Histology, University of the Basque Country UPV/EHU, Leioa 48940, Vizcaya, Spain
| | - F David Rodriguez
- Department of Biochemistry and Molecular Biology, E-37007, University of Salamanca, Salamanca, Spain
| | - Noelia Ruzafa
- Department of Cell Biology and Histology, University of the Basque Country UPV/EHU, Leioa 48940, Vizcaya, Spain
| | - Xandra Pereiro
- Department of Cell Biology and Histology, University of the Basque Country UPV/EHU, Leioa 48940, Vizcaya, Spain
| | - Sansar C Sharma
- Department of Ophthalmology, Cell Biology and Anatomy, New York Medical College, Valhalla, NY 10595, USA; IKERBASQUE, Basque Foundation for Science at Dept. Cell Biology and Histology, UPV/EHU, Spain
| |
Collapse
|
7
|
Soluble VCAM-1 impairs human brain endothelial barrier integrity via integrin α-4-transduced outside-in signalling. Acta Neuropathol 2015; 129:639-52. [PMID: 25814153 PMCID: PMC4405352 DOI: 10.1007/s00401-015-1417-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 03/10/2015] [Accepted: 03/21/2015] [Indexed: 01/17/2023]
Abstract
Human brain microvascular endothelial cells forming the blood–brain barrier (BBB) release soluble vascular cell adhesion molecule-1 (sVCAM-1) under inflammatory conditions. Furthermore, sVCAM-1 serum levels in untreated patients with multiple sclerosis (MS) correlate with a breakdown of the BBB as measured by gadolinium-enhanced MRI. To date, it is unknown whether sVCAM-1 itself modulates BBB permeability. Here, we provide evidence that human brain endothelium expresses integrin α-4/β-1, the molecular binding partner of sVCAM-1, and that sVCAM-1 directly impairs BBB function by inducing intracellular signalling events through integrin α-4. Primary human brain microvascular endothelial cells showed low to moderate integrin α-4 and strong β-1 but no definite β-7 expression in vitro and in situ. Increased brain endothelial integrin α-4 expression was observed in active MS lesions in situ and after angiogenic stimulation in vitro. Exposure of cultured primary brain endothelial cells to recombinant sVCAM-1 significantly increased their permeability to the soluble tracer dextran, which was paralleled by formation of actin stress fibres and reduced staining of tight junction-associated molecules. Soluble VCAM-1 was also found to activate Rho GTPase and p38 MAP kinase. Chemical inhibition of these signalling pathways partially prevented sVCAM-1-induced changes of tight junction arrangement. Importantly, natalizumab, a neutralising recombinant monoclonal antibody against integrin α-4 approved for the treatment of patients with relapsing–remitting MS, partially antagonised the barrier-disturbing effect of sVCAM-1. In summary, we newly characterised sVCAM-1 as a compromising factor of brain endothelial barrier function that may be partially blocked by the MS therapeutic natalizumab.
Collapse
|
8
|
Sakurai K, Matsuoka T, Suzuki C, Kinoshita J, Takayama G, Shimomura K. Investigation of the teratogenic potential of VLA-4 antagonist derivatives in rats. Reprod Toxicol 2014; 49:162-70. [PMID: 25194688 DOI: 10.1016/j.reprotox.2014.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 07/29/2014] [Accepted: 08/13/2014] [Indexed: 11/18/2022]
Abstract
Very late antigen-4 (VLA-4), which is concerned with cell-cell adhesion, plays important roles in development of the heart, and some VLA-4 antagonists cause cardiac anomalies. In this study, we evaluated the teratogenic potential of VLA-4 antagonist derivatives as screening, and investigated the conditions that induce cardiac anomalies. Seventeen compounds were orally administered to pregnant rats throughout the organogenesis period, and fetal examinations were performed. In addition, drug concentrations in the embryos were assayed. As a result, the incidence of ventricular septal defect (VSD) ranged from 0 to 100% depending on the compound. Plasma drug concentrations in the dams were related to increased incidence of VSD; however, these incidences were not increased when the concentration of the compound in the embryos at 24h after dosing was low. It is considered that continuous pharmacological activity in the embryo for more than 24h might disrupt closure of the ventricular septum.
Collapse
Affiliation(s)
- Ken Sakurai
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 16-13, Kita-Kasai 1-Chome, Edogawa-Ku, Tokyo 134-8630, Japan.
| | - Toshiki Matsuoka
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 16-13, Kita-Kasai 1-Chome, Edogawa-Ku, Tokyo 134-8630, Japan
| | - Chiharu Suzuki
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 16-13, Kita-Kasai 1-Chome, Edogawa-Ku, Tokyo 134-8630, Japan
| | - Junzo Kinoshita
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 16-13, Kita-Kasai 1-Chome, Edogawa-Ku, Tokyo 134-8630, Japan
| | - Gensuke Takayama
- Oncology Research Laboratories, Daiichi Sankyo Co., Ltd., 2-58, Hiromachi 1-Chome, Shinagawa-Ku, Tokyo 140-0005, Japan
| | - Kazuhiro Shimomura
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., 16-13, Kita-Kasai 1-Chome, Edogawa-Ku, Tokyo 134-8630, Japan
| |
Collapse
|
9
|
Wang W, Guo L, Yu Y, Chen Z, Zhou R, Yuan Z. Peptide REDV-modified polysaccharide hydrogel with endothelial cell selectivity for the promotion of angiogenesis. J Biomed Mater Res A 2014; 103:1703-12. [PMID: 25103847 DOI: 10.1002/jbm.a.35306] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 07/28/2014] [Indexed: 11/06/2022]
Abstract
Rapid and controlled vascularization of engineered tissues remains one of the key limitations in thick tissue engineering. Although many studies have focused on improving the rapid vascularization through the immobilization of bioactive molecules, the competition in growth between endothelial cells (ECs) and other cell types is to some extent neglected. In this study, we developed a peptide GREDV-modified scaffold for selective adhesion of human umbilical vein endothelial cells (HUVECs) through the specific recognition of the REDV peptide and integrin α4 β1 . In vitro studies showed that GREDV-conjugated alginate (ALG-GREDV) improved HUVEC adhesion, migration and proliferation when compared with a non-modified group. Furthermore, ALG-GREDV exhibited a superior capability for promoting the proliferation and selective adhesion of HUVEC over that of other peptide (RGD and YIGSR) modified groups (ALG-Pep). In vivo angiogenic assays demonstrated that the ALG-GREDV scaffold induced an angiogenic potential by stimulating new vessel formation and showed the highest blood vessel density among all samples after 21 days of implanting (83.7 vessels/mm(2) ). More importantly, the blood vessel density in cambium fibrous tissue of ALG-GREDV was about 1.5 times greater than other ALG-Pep groups, indicating facilitation of ALG-GREDV on selective angiogenesis in vivo. These results demonstrated that REDV-conjugated alginate could be a useful scaffold for stimulating and inducing angiogenesis in tissue-engineered applications.
Collapse
Affiliation(s)
- Wei Wang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, Nankai University, and Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin, 300071, China
| | | | | | | | | | | |
Collapse
|
10
|
Sturgeon CM, Chicha L, Ditadi A, Zhou Q, McGrath KE, Palis J, Hammond SM, Wang S, Olson EN, Keller G. Primitive erythropoiesis is regulated by miR-126 via nonhematopoietic Vcam-1+ cells. Dev Cell 2012; 23:45-57. [PMID: 22749417 DOI: 10.1016/j.devcel.2012.05.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Revised: 03/27/2012] [Accepted: 05/29/2012] [Indexed: 02/06/2023]
Abstract
Primitive erythropoiesis defines the onset of hematopoiesis in the yolk sac of the early embryo and is initiated by the emergence of progenitors assayed as colony-forming cells (EryP-CFCs). EryP-CFCs are detected for only a narrow window during embryonic development, suggesting that both their initiation and termination are tightly controlled. Using the embryonic stem differentiation system to model primitive erythropoiesis, we found that miR-126 regulates the termination of EryP-CFC development. Analyses of miR-126 null embryos revealed that this miR also regulates EryP-CFCs in vivo. We identified vascular cell adhesion molecule-1 (Vcam-1) expressed by a mesenchymal cell population as a relevant target of miR-126. Interaction of EryP-CFCs with Vcam-1 accelerated their maturation to ßh1-globin(+) and Ter119(+) cells through a Src family kinase. These findings uncover a cell nonautonomous regulatory pathway for primitive erythropoiesis that may provide insight into the mechanism(s) controlling the developmental switch from primitive to definitive hematopoiesis.
Collapse
|
11
|
Vincentz JW, Barnes RM, Firulli AB. Hand factors as regulators of cardiac morphogenesis and implications for congenital heart defects. BIRTH DEFECTS RESEARCH. PART A, CLINICAL AND MOLECULAR TERATOLOGY 2011; 91:485-94. [PMID: 21462297 PMCID: PMC3119928 DOI: 10.1002/bdra.20796] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Revised: 01/06/2011] [Accepted: 02/02/2011] [Indexed: 11/08/2022]
Abstract
Almost 15 years of careful study have established the related basic Helix-Loop-Helix (bHLH) transcription factors Hand1 and Hand2 as critical for heart development across evolution. Hand factors make broad contributions, revealed through animal models, to the development of multiple cellular lineages that ultimately contribute to the heart. They perform critical roles in ventricular cardiomyocyte growth, differentiation, morphogenesis, and conduction. They are also important for the proper development of the cardiac outflow tract, epicardium, and endocardium. Molecularly, they function both through DNA binding and through protein-protein interactions, which are regulated transcriptionally, posttranscriptionally by microRNAs, and posttranslationally through phosphoregulation. Although direct Hand factor transcriptional targets are progressively being identified, confirmed direct targets of Hand factor transcriptional activity in the heart are limited. Identification of these targets will be critical to model the mechanisms by which Hand factor bHLH interactions affect developmental pathways. Improved understanding of Hand factor-mediated transcriptional cascades will be necessary to determine how Hand factor dysregulation translates to human disease phenotypes. This review summarizes the insight that animal models have provided into the regulation and function of these factors during heart development, in addition to the recent findings that suggest roles for HAND1 and HAND2 in human congenital heart disease.
Collapse
Affiliation(s)
- Joshua W. Vincentz
- Riley Heart Research Center, Wells Center for Pediatric Research, Division of Pediatric Cardiology, Departments of Anatomy, Biochemistry and Medical and Molecular Genetics, Indiana Medical School, 1044 W. Walnut St., Indianapolis, IN 46202-5225, USA
| | - Ralston M. Barnes
- Riley Heart Research Center, Wells Center for Pediatric Research, Division of Pediatric Cardiology, Departments of Anatomy, Biochemistry and Medical and Molecular Genetics, Indiana Medical School, 1044 W. Walnut St., Indianapolis, IN 46202-5225, USA
| | - Anthony B. Firulli
- Riley Heart Research Center, Wells Center for Pediatric Research, Division of Pediatric Cardiology, Departments of Anatomy, Biochemistry and Medical and Molecular Genetics, Indiana Medical School, 1044 W. Walnut St., Indianapolis, IN 46202-5225, USA
| |
Collapse
|
12
|
Kuo BR, Erickson CA. Regional differences in neural crest morphogenesis. Cell Adh Migr 2010; 4:567-85. [PMID: 20962585 PMCID: PMC3011260 DOI: 10.4161/cam.4.4.12890] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Accepted: 07/02/2010] [Indexed: 12/11/2022] Open
Abstract
Neural crest cells are pluripotent cells that emerge from the neural epithelium, migrate extensively, and differentiate into numerous derivatives, including neurons, glial cells, pigment cells and connective tissue. Major questions concerning their morphogenesis include: 1) what establishes the pathways of migration and 2) what controls the final destination and differentiation of various neural crest subpopulations. These questions will be addressed in this review. Neural crest cells from the trunk level have been explored most extensively. Studies show that melanoblasts are specified shortly after they depart from the neural tube, and this specification directs their migration into the dorsolateral pathway. We also consider other reports that present strong evidence for ventrally migrating neural crest cells being similarly fate restricted. Cranial neural crest cells have been less analyzed in this regard but the preponderance of evidence indicates that either the cranial neural crest cells are not fate-restricted, or are extremely plastic in their developmental capability and that specification does not control pathfinding. Thus, the guidance mechanisms that control cranial neural crest migration and their behavior vary significantly from the trunk. The vagal neural crest arises at the axial level between the cranial and trunk neural crest and represents a transitional cell population between the head and trunk neural crest. We summarize new data to support this claim. In particular, we show that: 1) the vagal-level neural crest cells exhibit modest developmental bias; 2) there are differences in the migratory behavior between the anterior and the posterior vagal neural crest cells reminiscent of the cranial and the trunk neural crest, respectively; 3) the vagal neural crest cells take the dorsolateral pathway to the pharyngeal arches and the heart, but the ventral pathway to the peripheral nervous system and the gut. However, these pathways are not rigidly specified because of prior fate restriction. Understanding the molecular, cellular and behavioral differences between these three populations of neural crest cells will be of enormous assistance when trying to understand the evolution of the neck.
Collapse
Affiliation(s)
- Bryan R Kuo
- Department of Molecular and Cellular Biology, University of California, Davis, CA, USA
| | | |
Collapse
|
13
|
Holler KL, Hendershot TJ, Troy SE, Vincentz JW, Firulli AB, Howard MJ. Targeted deletion of Hand2 in cardiac neural crest-derived cells influences cardiac gene expression and outflow tract development. Dev Biol 2010; 341:291-304. [PMID: 20144608 DOI: 10.1016/j.ydbio.2010.02.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2009] [Revised: 01/29/2010] [Accepted: 02/01/2010] [Indexed: 11/29/2022]
Abstract
The basic helix-loop-helix DNA binding protein Hand2 has critical functions in cardiac development both in neural crest-derived and mesoderm-derived structures. Targeted deletion of Hand2 in the neural crest has allowed us to genetically dissect Hand2-dependent defects specifically in outflow tract and cardiac cushion independent of Hand2 functions in mesoderm-derived structures. Targeted deletion of Hand2 in the neural crest results in misalignment of the aortic arch arteries and outflow tract, contributing to development of double outlet right ventricle (DORV) and ventricular septal defects (VSD). These neural crest-derived developmental anomalies are associated with altered expression of Hand2-target genes we have identified by gene profiling. A number of Hand2 direct target genes have been identified using ChIP and ChIP-on-chip analyses. We have identified and validated a number of genes related to cell migration, proliferation/cell cycle and intracellular signaling whose expression is affected by Hand2 deletion in the neural crest and which are associated with development of VSD and DORV. Our data suggest that Hand2 is a multifunctional DNA binding protein affecting expression of target genes associated with a number of functional interactions in neural crest-derived cells required for proper patterning of the outflow tract, generation of the appropriate number of neural crest-derived cells for elongation of the conotruncus and cardiac cushion organization. Our genetic model has made it possible to investigate the molecular genetics of neural crest contributions to outflow tract morphogenesis and cell differentiation.
Collapse
Affiliation(s)
- Kristen L Holler
- Department of Neurosciences and Program in Neurosciences and Degenerative Disease, Health Sciences Campus, University of Toledo, 3000 Arlington Ave., Toledo, OH 43614-1007, USA
| | | | | | | | | | | |
Collapse
|
14
|
Abstract
Cardiac fibroblasts are the most populous nonmyocyte cell type within the mature heart and are required for extracellular matrix synthesis and deposition, generation of the cardiac skeleton, and to electrically insulate the atria from the ventricles. Significantly, cardiac fibroblasts have also been shown to play an important role in cardiomyocyte growth and expansion of the ventricular chambers during heart development. Although there are currently no cardiac fibroblast-restricted molecular markers, it is generally envisaged that the majority of the cardiac fibroblasts are derived from the proepicardium via epithelial-to-mesenchymal transformation. However, still relatively little is known about when and where the cardiac fibroblasts cells are generated, the lineage of each cell, and how cardiac fibroblasts move to reside in their final position throughout all four cardiac chambers. In this review, we summarize the present understanding regarding the function of Periostin, a useful marker of the noncardiomyocyte lineages, and its role during cardiac morphogenesis. Characterization of the cardiac fibroblast lineage and identification of the signals that maintain, expand and regulate their differentiation will be required to improve our understanding of cardiac function in both normal and pathophysiological states.
Collapse
Affiliation(s)
| | | | | | - Mohamad Azhar
- BIO5 Institute, University of Arizona, Tucson, AZ 85724
| | | | - Simon J. Conway
- Address for correspondence: Simon J. Conway, 1044 West Walnut Street, Room R4 W379, Indiana University School of Medicine, Indianapolis, IN 46202, USA. phone: (317) 278-8781; fax: (317) 278-5413;
| |
Collapse
|
15
|
Wehner NG, Shopp G, Osterburg I, Fuchs A, Buse E, Clarke J. Postnatal development in cynomolgus monkeys following prenatal exposure to natalizumab, an alpha4 integrin inhibitor. ACTA ACUST UNITED AC 2009; 86:144-56. [PMID: 19283860 DOI: 10.1002/bdrb.20193] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Natalizumab is a humanized monoclonal IgG4 antibody to human alpha4 integrin that blocks the interaction of alpha4beta1 and alpha4beta7 integrins with their ligands, including fibronectin, vascular cell adhesion molecule-1, and mucosal addressin cellular adhesion molecule-1. Because alpha4 integrins and their ligands are widely involved in mammalian development, lymphopoeisis, and hematopoiesis, natalizumab may interfere with these processes. METHODS The effects of prenatal exposure to natalizumab on postnatal development were assessed in cynomolgus monkeys at doses of 0 and 30 mg/kg administered intravenously every other day from gestational day (GD) 20 to 70 or GD 20 to term. Infants were delivered by natural birth and evaluated for general health, survival, development, and immunological structure and function at 12 or 18 months. RESULTS An increase in abortions was seen in the first cohort of natalizumab-treated dams (39.3 vs. 7.1% in the controls) but not in the second cohort (33.3, 37.5%). Infants in the term treatment group had elevated lymphocyte ( approximately 150%) and nucleated red blood cell counts ( approximately 400%), consistent with the pharmacological effect of natalizumab, and reductions in platelet counts ( approximately 28%), which were reversible following clearance of natalizumab. No anemia was observed. Infants in the term treatment group had significantly increased spleen weights at 12 months but not at 18 months. All other experimental observations in infants from natalizumab-treated dams were comparable with those of controls. CONCLUSION Natalizumab had no adverse effects on the general health, survival, development, or immunological structure and function of infants born to dams treated with natalizumab during pregnancy.
Collapse
Affiliation(s)
- Nancy G Wehner
- Elan Pharmaceuticals, Inc., South San Francisco, California 94080, USA.
| | | | | | | | | | | |
Collapse
|
16
|
Wehner NG, Skov M, Shopp G, Rocca MS, Clarke J. Effects of natalizumab, an α4 integrin inhibitor, on fertility in male and female guinea pigs. ACTA ACUST UNITED AC 2009; 86:108-16. [DOI: 10.1002/bdrb.20191] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
17
|
Wehner NG, Shopp G, Oneda S, Clarke J. Embryo/fetal development in cynomolgus monkeys exposed to natalizumab, an α4 integrin inhibitor. ACTA ACUST UNITED AC 2009; 86:117-30. [DOI: 10.1002/bdrb.20190] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
18
|
Wehner NG, Shopp G, Rocca MS, Clarke J. Effects of natalizumab, an α4 integrin inhibitor, on the development of Hartley guinea pigs. ACTA ACUST UNITED AC 2009; 86:98-107. [DOI: 10.1002/bdrb.20189] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
19
|
Astrof S, Hynes RO. Fibronectins in vascular morphogenesis. Angiogenesis 2009; 12:165-75. [PMID: 19219555 DOI: 10.1007/s10456-009-9136-6] [Citation(s) in RCA: 186] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2009] [Accepted: 01/30/2009] [Indexed: 11/28/2022]
Abstract
Fibronectin is an extracellular matrix protein found only in vertebrate organisms containing endothelium-lined vasculature and is required for cardiovascular development in fish and mice. Fibronectin and its splice variants containing EIIIA and EIIIB domains are highly upregulated around newly developing vasculature during embryogenesis and in pathological conditions including atherosclerosis, cardiac hypertrophy, and tumorigenesis. However, their molecular roles in these processes are not entirely understood. We review genetic studies examining functions of fibronectin and its splice variants during embryonic cardiovascular development, and discuss potential roles of fibronectin in vascular disease and tumor angiogenesis.
Collapse
Affiliation(s)
- Sophie Astrof
- Greenberg Division of Cardiology, Department of Medicine, Weill Medical College of Cornell University, New York, NY 10065, USA.
| | | |
Collapse
|
20
|
Costa-Silva B, da Costa MC, Melo FR, Neves CM, Alvarez-Silva M, Calloni GW, Trentin AG. Fibronectin promotes differentiation of neural crest progenitors endowed with smooth muscle cell potential. Exp Cell Res 2009; 315:955-67. [PMID: 19331824 DOI: 10.1016/j.yexcr.2009.01.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2008] [Revised: 01/11/2009] [Accepted: 01/17/2009] [Indexed: 12/12/2022]
Abstract
The neural crest (NC) is a model system used to investigate multipotency during vertebrate development. Environmental factors control NC cell fate decisions. Despite the well-known influence of extracellular matrix molecules in NC cell migration, the issue of whether they also influence NC cell differentiation has not been addressed at the single cell level. By analyzing mass and clonal cultures of mouse cephalic and quail trunk NC cells, we show for the first time that fibronectin (FN) promotes differentiation into the smooth muscle cell phenotype without affecting differentiation into glia, neurons, and melanocytes. Time course analysis indicated that the FN-induced effect was not related to massive cell death or proliferation of smooth muscle cells. Finally, by comparing clonal cultures of quail trunk NC cells grown on FN and collagen type IV (CLIV), we found that FN strongly increased both NC cell survival and the proportion of unipotent and oligopotent NC progenitors endowed with smooth muscle potential. In contrast, melanocytic progenitors were prominent in clonogenic NC cells grown on CLIV. Taken together, these results show that FN promotes NC cell differentiation along the smooth muscle lineage, and therefore plays an important role in fate decisions of NC progenitor cells.
Collapse
Affiliation(s)
- Bruno Costa-Silva
- Departamento de Biologia Celular, Embriologia e Genética, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Campus Universitário-Trindade, 88040-900 Florianópolis, S.C., Brazil
| | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
Angiogenesis, the formation of new blood vessels from preexisting vasculature, contributes to the pathogenesis of many disorders, including ischemic diseases and cancer. Integrins are cell adhesion molecules that are expressed on the surface of endothelial cells and pericytes, making them potential targets for antiangiogenic therapy. Here we review the contribution of endothelial and mural cell integrins to angiogenesis and highlight their potential as antiangiogenesis targets.
Collapse
Affiliation(s)
- Rita Silva
- From the Adhesion and Angiogenesis Group, Centre for Tumour Biology, Cancer Research UK Clinical Centre and the Institute of Cancer, Barts & The London & Queen Mary’s School of Medicine & Dentistry, John Vane Science Centre, Charterhouse Square, London UK
| | - Gabriela D'Amico
- From the Adhesion and Angiogenesis Group, Centre for Tumour Biology, Cancer Research UK Clinical Centre and the Institute of Cancer, Barts & The London & Queen Mary’s School of Medicine & Dentistry, John Vane Science Centre, Charterhouse Square, London UK
| | - Kairbaan M. Hodivala-Dilke
- From the Adhesion and Angiogenesis Group, Centre for Tumour Biology, Cancer Research UK Clinical Centre and the Institute of Cancer, Barts & The London & Queen Mary’s School of Medicine & Dentistry, John Vane Science Centre, Charterhouse Square, London UK
| | - Louise E. Reynolds
- From the Adhesion and Angiogenesis Group, Centre for Tumour Biology, Cancer Research UK Clinical Centre and the Institute of Cancer, Barts & The London & Queen Mary’s School of Medicine & Dentistry, John Vane Science Centre, Charterhouse Square, London UK
| |
Collapse
|
22
|
Goldfinger LE, Tzima E, Stockton R, Kiosses WB, Kinbara K, Tkachenko E, Gutierrez E, Groisman A, Nguyen P, Chien S, Ginsberg MH. Localized alpha4 integrin phosphorylation directs shear stress-induced endothelial cell alignment. Circ Res 2008; 103:177-85. [PMID: 18583710 DOI: 10.1161/circresaha.108.176354] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Vascular endothelial cells respond to laminar shear stress by aligning in the direction of flow, a process which may contribute to atheroprotection. Here we report that localized alpha4 integrin phosphorylation is a mechanism for establishing the directionality of shear stress-induced alignment in microvascular endothelial cells. Within 5 minutes of exposure to a physiological level of shear stress, endothelial alpha4 integrins became phosphorylated on Ser(988). In wounded monolayers, phosphorylation was enhanced at the downstream edges of cells relative to the source of flow. The shear-induced alpha4 integrin phosphorylation was blocked by inhibitors of cAMP-dependent protein kinase A (PKA), an enzyme involved in the alignment of endothelial cells under prolonged shear. Moreover, shear-induced localized activation of the small GTPase Rac1, which specifies the directionality of endothelial alignment, was similarly blocked by PKA inhibitors. Furthermore, endothelial cells bearing a nonphosphorylatable alpha4(S(988)A) mutation failed to align in response to shear stress, thus establishing alpha4 as a relevant PKA substrate. We thereby show that shear-induced PKA-dependent alpha4 integrin phosphorylation at the downstream edge of endothelial cells promotes localized Rac1 activation, which in turn directs cytoskeletal alignment in response to shear stress.
Collapse
Affiliation(s)
- Lawrence E Goldfinger
- Sol Sherry Thrombosis Research Center and Department of Anatomy & Cell Biology, Temple University School of Medicine, 3400 N. Broad Street, OMS 415, Philadelphia, PA 19140, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Integrins in Angiogenesis. Angiogenesis 2008. [DOI: 10.1007/978-0-387-71518-6_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
24
|
Kwee L, Burns DK, Rumberger JM, Norton C, Wolitzky B, Terry R, Lombard-Gillooly KM, Shuster DJ, Kontgen F, Stewart C. Creation and characterization of E-selectin- and VCAM-1-deficient mice. CIBA FOUNDATION SYMPOSIUM 2007; 189:17-28; discussion 28-34, 77-8. [PMID: 7587631 DOI: 10.1002/9780470514719.ch3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
A variety of adhesion molecules have been identified which mediate the interaction of leukocytes with endothelial cells. In order to define the role of individual molecules in inflammation we have produced lines of mice which are deficient in the synthesis of specific adhesion molecules. Null mutations were introduced into the genes encoding E-selectin or vascular cell adhesion molecule-1 (VCAM-1) in embryonic stem cells and these cells were used to produce lines of mice carrying the mutation. E-selectin-deficient mice were viable and exhibited no developmental defects. The roles of E- and P-selectin in the influx of neutrophils were examined using these mice. The data suggest that the two selectins are functionally redundant in mediating neutrophil emigration in a model of chemically induced peritonitis. VCAM-1-deficient mice are not viable. Analysis of VCAM-1 gene expression in wild-type embryos and phenotypic analysis of VCAM-1 -/- embryos suggests that VCAM-1 is required for development of the extraembryonic circulatory system and the embryonic heart.
Collapse
Affiliation(s)
- L Kwee
- Roche Research Center, Department of Biotechnology, Hoffman-La Roche Inc., Nutley, NJ 07110-1199, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Caruso M, Busanello A, Sthandier O, Cavaldesi M, Gentile M, Garcia MI, Amati P. Mutation in the VP1-LDV motif of the murine polyomavirus affects viral infectivity and conditions virus tissue tropism in vivo. J Mol Biol 2006; 367:54-64. [PMID: 17239397 DOI: 10.1016/j.jmb.2006.12.059] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2006] [Revised: 12/18/2006] [Accepted: 12/19/2006] [Indexed: 11/24/2022]
Abstract
The first contact of a virus with the host cell surface and further entry are important steps for a successful outcome of the infection process and for the virus-associated pathogenicity. We have previously shown that the entry of the murine Polyomavirus (Py) into fibroblasts is a multi-step process involving, at least, the attachment to primary sialic acids (SA)-containing cell receptors followed by post-binding interaction with secondary receptors, such as the alpha4beta1 integrin, likely through the VP1-LDV motif. Here we report on the functional role of the VP1-LDV motif in Py infectivity and in vivo virus tissue tropism. For this purpose, we have characterized a recombinant virus mutant, PyLNV, harboring a single aa substitution in this motif (D138N). Although not critical for virus viability, the D138N substitution abrogates the post-attachment Py-alpha4beta1 interaction, rendering the PyLNV mutant virus twofold less infectious than the Py wild-type (Wt) in alpha4beta1-positive fibroblasts. To study the putative role of the VP1-LDV motif in vivo, newborn C57BL/6 mice were inoculated with PyWt or PyLNV and, after six days, organs were analyzed for the presence of viral DNA. Intriguingly, PyLNV showed an altered spectrum of in vivo replication compared with PyWt, particularly in the skin and in the kidney. The implication of Py-alpha4beta1 integrin interaction in conditioning tissue-specificity of virus replication is discussed.
Collapse
Affiliation(s)
- Maddalena Caruso
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Biotecnologie Cellulari ed Ematologia, Sezione di Genetica Molecolare, Università di Roma La Sapienza, Viale Regina Elena 324, 00161 Rome, Italy.
| | | | | | | | | | | | | |
Collapse
|
26
|
Shinoda G, Umeda K, Heike T, Arai M, Niwa A, Ma F, Suemori H, Luo HY, Chui DHK, Torii R, Shibuya M, Nakatsuji N, Nakahata T. alpha4-Integrin(+) endothelium derived from primate embryonic stem cells generates primitive and definitive hematopoietic cells. Blood 2006; 109:2406-15. [PMID: 17090646 DOI: 10.1182/blood-2006-06-031039] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The mechanism of commencement of hematopoiesis in blood islands of the yolk sac and the aorta-gonad-mesonephros (AGM) region during primate embryogenesis remains elusive. In this study, we demonstrated that VE-cadherin(+)CD45(-) endothelial cells derived from nonhuman primate embryonic stem cells are able to generate primitive and definitive hematopoietic cells sequentially, as revealed by immunostaining of floating erythrocytes and colony-forming assay in cultures. Single bipotential progenitors for hematopoietic and endothelial lineages are included in this endothelial cell population. Furthermore, hemogenic activity of these endothelial cells is observed exclusively in the alpha4-integrin(+) subpopulation; bipotential progenitors are 4-fold enriched in this subpopulation. The kinetics of this hemogenic subpopulation is similar to that of hemogenic endothelial cells previously reported in the yolk sac and the AGM region in vivo in that they emerge for only a limited time. We suggest that VE-cadherin(+)CD45(-)alpha4-integrin(+) endothelial cells are involved in primitive and definitive hematopoiesis during primate embryogenesis, though VE-cadherin(-)CD45(-)alpha4-integrin(+) cells are the primary sources for primitive hematopoiesis.
Collapse
Affiliation(s)
- Gen Shinoda
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Grazioli A, Alves CS, Konstantopoulos K, Yang JT. Defective blood vessel development and pericyte/pvSMC distribution in alpha 4 integrin-deficient mouse embryos. Dev Biol 2006; 293:165-77. [PMID: 16529735 DOI: 10.1016/j.ydbio.2006.01.026] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2005] [Revised: 01/20/2006] [Accepted: 01/27/2006] [Indexed: 11/16/2022]
Abstract
Blood vessel development is in part regulated by pericytes/presumptive vascular smooth muscle cells (PC/pvSMCs). Here, we demonstrate that interactions between PC/pvSMCs and extracellular matrix play a critical role in this event. We show that the cranial vessels in alpha4 integrin-deficient mouse embryos at the stage of vessel remodeling are increased in diameter. This defect is accompanied by a failure of PC/pvSMCs, which normally express alpha4beta1 integrin, to spread uniformly along the vessels. We also find that fibronectin but not VCAM-1 is localized in the cranial vessels at this stage. Furthermore, cultured alpha4 integrin-null PC/pvSMCs plated on fibronectin display a delay in initiating migration, a reduction in migration speed, and a decrease in directional persistence in response to a polarized force of shear flow. These results suggest that specific motile activities of PC/pvSMCs regulated by mechanical signals imposed by the interstitial extracellular matrix may also be required in vivo for the distribution and function of the PC/pvSMCs during blood vessel development.
Collapse
Affiliation(s)
- Alison Grazioli
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | |
Collapse
|
28
|
Abstract
The effects of growth factors on tissue remodeling and cell differentiation depend on the nature of the extracellular matrix, the type and organization of integrins, the activation of metalloproteinases and the presence of secreted proteins associated to the matrix. These interactions are actually poorly known in the cardiovascular system. We describe here: 1) the main components of extracellular matrix within the cardiovascular system; 2) the role of integrins in the transmission of growth signals; 3) the shift in the expression of the components of the extracellular matrix (fibronectin and collagens) and the stimulation of the synthesis of metalloproteinases during normal and hypertrophic growth of the myocardium; 4) the effects of growth factors, such as Angiotensin II, Fibroblast Growth Factors (FGF), Transforming Growth Factor-beta (TGF-beta), on the synthesis of proteins of the extracellular matrix in the heart.
Collapse
Affiliation(s)
- S Corda
- Hôpital Lariboisière, INSERM U 127, Paris, France
| | | | | |
Collapse
|
29
|
Wingerd KL, Wayne WC, Jackson DY, Clegg DO. Involvement of alpha4 integrins in maintenance of cardiac sympathetic axons. Auton Neurosci 2005; 122:58-68. [PMID: 16181811 DOI: 10.1016/j.autneu.2005.08.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2005] [Revised: 08/01/2005] [Accepted: 08/06/2005] [Indexed: 11/15/2022]
Abstract
Sympathetic neurons extend and maintain axons that innervate the myocardium, and proper innervation is important for cardiac function. However, the molecular basis for axon outgrowth and maintenance is not well understood. We have shown previously that the integrin alpha4beta1 is expressed on developing axons, and the alpha4 function is important for the development of innervation in vivo [Wingerd, K.L., Goodman, N.L., Tresser, J.W., Smail, M.M., Leu, S.T., Rohan, S.J., Pring, J.L., Jackson, D.Y., and Clegg, D.O., 2002. Alpha 4 integrins and vascular cell adhesion molecule-1 play a role in sympathetic innervation of the heart. J. Neurosci. 22,10772-10780]. Here we examine the function of alpha4beta1 integrins in the maintenance of cardiac sympathetic innervation in vitro and in vivo, and investigate integrin expression and function after myocardial infarction and in hypertensive rats. On substrates of vascular cell adhesion molecule-1 (VCAM-1), alpha4beta1 was required for both initial outgrowth and maintenance of neurites in vitro. On fibronectin substrates, initial outgrowth requires only alpha4 integrins, but maintenance requires both alpha4 integrins and RGD-dependent integrins. In vivo, in adult Long Evans rats, inhibition of alpha4 integrins resulted in decreased maintenance of sympathetic fibers innervating the apex of the heart. However, alpha4 integrins were not detected on most sympathetic axons that sprout after myocardial infarction, and alpha4 function was not required for sprouting. Spontaneously hypertensive rats (SHR) have increased numbers of cardiac sympathetic fibers compared to the parental Wistar strain, but many of these lack alpha4 expression, and alpha4 function is not required for maintenance of these fibers in the heart. These results suggest that developing sympathetic axons and sprouting sympathetic axons use different mechanisms of outgrowth, and that maintenance of cardiac sympathetic innervation involves alpha4 integrins in some rat strains.
Collapse
Affiliation(s)
- Kevin L Wingerd
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA
| | | | | | | |
Collapse
|
30
|
Williams SS, Mear JP, Liang HC, Potter SS, Aronow BJ, Colbert MC. Large-scale reprogramming of cranial neural crest gene expression by retinoic acid exposure. Physiol Genomics 2005; 19:184-97. [PMID: 15466718 DOI: 10.1152/physiolgenomics.00136.2004] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Although retinoic acid (RA), the active form of vitamin A, is required for normal embryonic growth and development, it is also a powerful teratogen. Infants born to mothers exposed to retinoids during pregnancy have a 25-fold increased risk for malformations, nearly exclusively of cranial neural crest-derived tissues. To characterize neural crest cell responses to RA, we exposed murine crest cultures to teratogenic levels of RA and subjected their RNA to microarray-based gene expression profile analysis using Affymetrix MG-U74Av2 GeneChips. RNAs were isolated from independent cultures treated with 10(-6) M RA for 6, 12, 24, or 48 h. Statistical analyses of gene expression profile data facilitated identification of the 205 top-ranked differentially regulated genes whose expression was reproducibly changed by RA over time. Cluster analyses of these genes across the independently treated sample series revealed distinctive kinetic patterns of altered gene expression. The largest group was transiently affected within the first 6 h of exposure, representing early responding genes. Group 2 showed sustained induction by RA over all times, whereas group 3 was characterized by the suppression of a time-dependent expression increase normally seen in untreated cells. Additional patterns demonstrated time-dependent increased or decreased expression among genes not normally regulated to a significant extent. Gene function analysis revealed that more than one-third of all RA-regulated genes were associated with developmental regulation, including both canonical and noncanonical Wnt signaling pathways. Multiple genes associated with cell adhesion and cell cycle regulation, recognized targets for the biological effects of RA, were also affected. Taken together, these results support the hypothesis that the teratogenic effects of RA derive from reprogramming gene expression of a host of genes, which play critical roles during embryonic development regulating pathways that determine subsequent differentiation of cranial neural crest cells.
Collapse
Affiliation(s)
- Sarah S Williams
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA
| | | | | | | | | | | |
Collapse
|
31
|
Abstract
The growth of new blood vessels is a dynamic yet highly regulated process that depends on coordinated signaling by growth factor and cell adhesion receptors. As part of the molecular program regulating angiogenesis, endothelial cells acquire a proliferative and invasive phenotype but also show increased susceptibility to apoptotic stimuli. Integrins are the principle adhesion receptors used by endothelial cells to interact with their extracellular microenvironment, and integrin-mediated interactions play a critical role in regulating cell proliferation, migration, and survival. Alterations in the repertoire and?or activity of integrins, as well as the availability and structural property of their ligands, regulate the vascular cell during the growth or repair of blood vessels.
Collapse
Affiliation(s)
- D G Stupack
- Department of Immunology, The Scripps Research Institute, La Jolla, California 9203, USA
| | | |
Collapse
|
32
|
Crofts F, Rohatagi S, Pino M, DeLise B, Zhang J, Nguyen M, Guittin P, Barbellion S, Brunel P, Hofmann T, Schmidt J, Wong M, Lockey P, Lerman S, Clark R. Critical period for a teratogenic VLA-4 antagonist: Developmental effects and comparison of embryo drug concentrations of teratogenic and non-teratogenic VLA-4 antagonists. ACTA ACUST UNITED AC 2004; 71:69-79. [PMID: 15098200 DOI: 10.1002/bdrb.20000] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Integrins such as VLA-4 (Very late antigen 4, integrin alpha4beta1) play key roles in cell-cell interactions that are critical for development. Homozygous null knockouts of the VLA-4 alpha4-subunit or VCAM-1 (VLA-4 cell surface ligand) in mice result in failure of the allantois and chorion to fuse leading to interrupted placentation and cardiac development and embryo lethality. Embryo-fetal studies of three VLA-4 antagonists, IVL745, IVL984, and HMR1031 [Crofts et al., Birth Defects Res B 71:55-68 (this issue), 2004] with exposure on gestation days (GD) 6-17 (rat), 6-18 (rabbit) or 6-15 (mouse) showed that only IVL984 treatment resulted in embryo lethality and cardiac defects. Objectives of the current study were to determine the critical period for inducing IVL984-related embryo-fetal effects, and to test the hypothesis that these effects were due to higher embryo drug concentrations. METHODS IVL984 was administered at 40 mg/kg/day to pregnant rats on GD 4 and 5, GD 6 and 7, GD 8 and 9, GD 10 and 11, or GD 12 and 13. Animals were euthanized on GD 21 and uteri and fetuses were examined. A treatment period of GD 10-12 was selected for subsequent toxicokinetic (TK) studies in which IVL984, HMR1031, or IVL745 was administered to pregnant rats and rabbits. On GD 12, maternal plasma, extra-embryonic tissue (placenta and amniotic fluid), and embryonic tissue were collected and analyzed for drug concentrations. RESULTS In the IVL984 critical period study in pregnant rats, treatment on GD 10 and 11 resulted in increased post-implantation loss, skeletal variations, and spiral septal defects similar to those observed in standard embryo-fetal development studies with treatment throughout organogenesis. There were no embryo-fetal effects after treatment on GD 4 and 5, GD 6 and 7, or GD 8 and 9. There was a single aorta malformation after treatment on GD 12 and 13. In the TK studies, IVL745, HMR1031, and IVL984 were all detectable in embryonic tissue and there was no evidence for accumulation. Rat and rabbit embryo exposures (AUC or dose-adjusted AUC) on GD 12 could not explain the observed teratology (IVL984<HMR1031<IVL745). Further analyses incorporating pharmacological activity, clearance, and protein binding data provided a positive correlation between embryonic exposure and teratogenic potency. CONCLUSIONS The critical period for IVL984 in the rat, GD 10 to 11, corresponds to the expression of alpha-4 integrin on the chorion and VCAM-1 on the allantois and myocardium as well as chorioallantoic fusion and formation of the spiral septum. Embryo drug levels adjusted for pharmacological activity, clearance, and protein binding provide a possible explanation for the differing teratogenic potency of IVL984, HMR1031, and IVL745.
Collapse
Affiliation(s)
- F Crofts
- Department of Drug Safety Evaluation, Aventis Inc., Bridgewater, New Jersey 08807, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Leu ST, Jacques SAL, Wingerd KL, Hikita ST, Tolhurst EC, Pring JL, Wiswell D, Kinney L, Goodman NL, Jackson DY, Clegg DO. Integrin α4β1 function is required for cell survival in developing retina. Dev Biol 2004; 276:416-30. [PMID: 15581875 DOI: 10.1016/j.ydbio.2004.09.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2004] [Revised: 08/31/2004] [Accepted: 09/01/2004] [Indexed: 11/24/2022]
Abstract
In the retina, integrins in the beta1 family have been shown to be important in many phases of neuronal development, particularly neuroblast migration and axon outgrowth. However, the functions of specific integrin heterodimers are not well defined. In this study, we investigated the functions of beta1 integrins in developing chicken retina by expression of a dominant-negative beta1A construct using a replication-competent retrovirus. Inhibition of integrins using this approach resulted in alteration of cell morphology and increased apoptosis, but did not preclude migration and axon elongation. In an attempt to identify which specific beta1 heterodimer was important, expression and function of the alpha4beta1 heterodimer were also investigated. At early developmental stages, alpha4 protein and mRNA were detected in undifferentiated neuroblasts throughout the retina. At later stages, expression was confined to retinal ganglion cells (RGCs) and amacrine cells. A small molecule antagonist of alpha4 integrins was shown to inhibit neurite outgrowth on recombinant soluble vascular cell adhesion molecule-1 (VCAM-1), a known ligand of alpha4beta1. Introduction of alpha4 antagonist in vivo gave rise to increased apoptosis and led to a thinning of the retina and reduced numbers of retinal ganglion cells (RGCs). We conclude that the integrin alpha4beta1 is important for survival of developing retinal neurons, including RGCs.
Collapse
Affiliation(s)
- Sergiu T Leu
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Wingerd KL, Goodman NL, Leu ST, Clegg DO. Expression and function of integrin ?4?1 and vascular cell adhesion molecule-1 (VCAM-1) during sympathetic innervation of the heart. Dev Dyn 2004; 231:359-69. [PMID: 15366013 DOI: 10.1002/dvdy.20120] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The interaction between the integrin alpha4beta1 receptor on superior cervical ganglion (SCG) neurons and vascular cell adhesion molecule-1 (VCAM-1) in cardiac tissue has been implicated in proper development of the sympathetic innervation of the heart (Wingerd et al. [2002] J Neurosci 22:10772-10780). In this study, we examined the expression and function of alpha4beta1 and VCAM-1 in developing rat SCG and heart. In vitro, the alpha4beta1-dependent neurite outgrowth on VCAM-1 decreased by approximately 50% from postnatal day 1 to 6. This down-regulation was correlated with a shift in alpha4 isoform and a shift in alpha4 localization from neurites to cell bodies. This altered localization was also observed in vivo but on a different time scale. alpha4 was detected on most developing SCG neurons and on macrophages and blood vessels. In the heart, alpha4 was detected on sympathetic axons, but the percentage of alpha4-positive fibers decreased with age. VCAM-1 immunoreactivity was abundant in heart tissue throughout development, in close proximity to sympathetic axons. The regulation of alpha4beta1 function, and localization of alpha4 and VCAM-1, are consistent with a role for the alpha4beta1--VCAM-1 interaction in extension of sympathetic axons into the myocardium.
Collapse
Affiliation(s)
- Kevin L Wingerd
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, California 93106, USA
| | | | | | | |
Collapse
|
35
|
Crofts F, Pino M, DeLise B, Guittin P, Barbellion S, Brunel P, Potdevin S, Bergmann B, Hofmann T, Lerman S, Clark RL. Different embryo-fetal toxicity effects for three VLA-4 antagonists. ACTA ACUST UNITED AC 2004; 71:55-68. [PMID: 15098199 DOI: 10.1002/bdrb.20001] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND VLA-4 (Very late antigen 4, integrin alpha4beta1) plays an important role in cell-cell interactions that are critical for development. Homozygous null knockouts of the alpha4 subunit of VLA-4 or VCAM-1 (cell surface ligand to VLA-4) in mice result in abnormal placental and cardiac development and embryo lethality. Objectives of the current study were to assess and compare the teratogenic potential of three VLA-4 antagonists. METHODS IVL745, HMR1031, and IVL984 were each evaluated by the subcutaneous route in standard embryo-fetal developmental toxicity studies in rats and rabbits. IVL984 was also evaluated in mice. Fetuses were examined externally, viscerally, and skeletally. RESULTS IVL745 did not cause significant maternal or fetal effects at doses up to 100 or 250 mg/kg/day in rats or rabbits, respectively. HMR1031 treatment resulted in marked maternal toxicity and slight fetal toxicity at the highest tested doses of 200 and 75 mg/kg/day in rats and rabbits, respectively. HMR1031 embryo-fetal effects consisted of slightly lower body weight and crown-rump length in rats and minor sternebral defects in rabbits. IVL984 treatment resulted in minimal maternal effects at doses up to 40, 15, and 100 mg/kg/day in rats, rabbits, and mice, respectively (excluding abortions in rabbits). However, marked developmental effects were observed at the lowest tested IVL984 doses, 1, 0.2, and 3 mg/kg/day in rats, rabbits, and mice, respectively. IVL984 embryo-fetal effects consisted of increased total post-implantation loss due to early resorptions and high incidences of cardiac malformations and skeletal malformations and/or variations. Notably, spiral septal defects were observed in up to 76% of rat fetuses and up to 58% of rabbit fetuses. CONCLUSIONS Dramatic differences in teratogenic potential were observed: IVL745 was not teratogenic, HMR1031 caused slight embryo-fetal effects at maternally-toxic doses, and IVL984 was a potent teratogen at doses where direct maternal toxicity was limited to abortions in rabbits. Prominent effects of IVL984 included embryo lethality and cardiac malformations including spiral septal defects in three species.
Collapse
Affiliation(s)
- F Crofts
- Department of Drug Safety Evaluation, Aventis Inc., Bridgewater, New Jersey 08807, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Calzada MJ, Zhou L, Sipes JM, Zhang J, Krutzsch HC, Iruela-Arispe ML, Annis DS, Mosher DF, Roberts DD. Alpha4beta1 integrin mediates selective endothelial cell responses to thrombospondins 1 and 2 in vitro and modulates angiogenesis in vivo. Circ Res 2003; 94:462-70. [PMID: 14699013 DOI: 10.1161/01.res.0000115555.05668.93] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We examined the function of alpha4beta1 integrin in angiogenesis and in mediating endothelial cell responses to the angiogenesis modulators, thrombospondin-1 and thrombospondin-2. Alpha4beta1 supports adhesion of venous endothelial cells but not of microvascular endothelial cells on immobilized thrombospondin-1, vascular cell adhesion molecule-1, or recombinant N-terminal regions of thrombospondin-1 and thrombospondin-2. Chemotactic activities of this region of thrombospondin-1 and thrombospondin-2 are also mediated by alpha4beta1, whereas antagonism of fibroblast growth factor-2-stimulated chemotaxis is not mediated by this region. Immobilized N-terminal regions of thrombospondin-1 and thrombospondin-2 promote endothelial cell survival and proliferation in an alpha4beta1-dependent manner. Soluble alpha4beta1 antagonists inhibit angiogenesis in the chick chorioallantoic membrane and neovascularization of mouse muscle explants. The latter inhibition is thrombospondin-1-dependent and not observed in explants from thrombospondin-1-/- mice. Antagonizing alpha4beta1 may in part block proangiogenic activities of thrombospondin-1 and thrombospondin-2, because N-terminal regions of thrombospondin-1 and thrombospondin-2 containing the alpha4beta1 binding sequence stimulate angiogenesis in vivo. Therefore, alpha4beta1 is an important endothelial cell receptor for mediating motility and proliferative responses to thrombospondins and for modulation of angiogenesis.
Collapse
Affiliation(s)
- Maria J Calzada
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, Md 20892-1500, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Hosking BM, Wang SCM, Downes M, Koopman P, Muscat GEO. The VCAM-1 gene that encodes the vascular cell adhesion molecule is a target of the Sry-related high mobility group box gene, Sox18. J Biol Chem 2003; 279:5314-22. [PMID: 14634005 DOI: 10.1074/jbc.m308512200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
VCAM-1 (vascular cell adhesion molecule-1) and Sox18 are involved in vascular development. VCAM-1 is an important adhesion molecule that is expressed on endothelial cells and has a critical role in endothelial activation, inflammation, lymphatic pathophysiology, and atherogenesis. The Sry-related high mobility group box factor Sox18 has previously been implicated in endothelial pathologies. Mutations in human and mouse Sox18 leads to hypotrichosis and lymphedema. Furthermore, both Sox18 and VCAM-1 have very similar spatio-temporal patterns of expression, which is suggestive of cross-talk. We use biochemical techniques, cell culture systems, and the ragged opossum (RaOP) mouse model with a naturally occurring mutation in Sox18 to demonstrate that VCAM-1 is an important target of Sox18. Transfection, site-specific mutagenesis, and gel shift analyses demonstrated that Sox18 directly targeted and trans-activated VCAM-1 expression. Importantly, the naturally occurring Sox18 mutant attenuates the expression and activation of VCAM-1 in vitro. Furthermore, in vivo quantitation of VCAM-1 mRNA levels in wild type and RaOP mice demonstrates that RaOP animals show a dramatic and significant reduction in VCAM-1 mRNA expression in lung, skin, and skeletal muscle. Our observation that the VCAM-1 gene is an important target of SOX18 provides the first molecular insights into the vascular abnormalities in the mouse mutant ragged and the human hypotrichosis-lymphedema-telangiectasia disorder.
Collapse
Affiliation(s)
- Brett M Hosking
- Institute for Molecular Bioscience, Queensland Biosciences Precinct, University of Queensland, Brisbane, Queensland 4072, Australia
| | | | | | | | | |
Collapse
|
38
|
Hikita ST, Cann GM, Wingerd KL, Mullick LH, Wayne WC, Webb SW, Clegg DO. Integrin alpha4beta1 (VLA-4) expression and activity in retinal and peripheral neurons. Mol Cell Neurosci 2003; 23:427-39. [PMID: 12837626 DOI: 10.1016/s1044-7431(03)00065-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The integrin alpha4beta1 fulfills important roles in inflammation and hematopoesis, but its functions in neurons are not well understood. Here we show that the alpha4 subunit is expressed on mouse retinal ganglion cells (RGCs) and undifferentiated retinal neuroblasts during the period of axon extension and migration. To determine if alpha4 integrins expressed by retinal neurons were active, neurons were cultured on known alpha4 ligands in vitro. Recombinant soluble vascular cell adhesion molecule 1 (rsVCAM-1), fibronectin, and osteopontin (OPN) induced neurite outgrowth that was diminished by function blocking antibodies specific for alpha4. Neurite outgrowth on OPN was also blocked by antibodies to the integrin beta1 subunit, implicating the alpha4beta1 heterodimer as one integrin receptor mediating outgrowth on OPN. OPN immunoreactivity was detected in the RGC fiber layer and optic nerve, suggesting that it may act as an alpha4 ligand in vivo. Neurons from chick lumbar sympathetic ganglia, chick dorsal root ganglia, and mouse superior cervical ganglia also extended neurites on rsVCAM-1, suggesting that integrin alpha4beta1 may play a role in the development of multiple neuronal cell types.
Collapse
Affiliation(s)
- Sherry T Hikita
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Alpha 4 integrins and vascular cell adhesion molecule-1 play a role in sympathetic innervation of the heart. J Neurosci 2003. [PMID: 12486170 DOI: 10.1523/jneurosci.22-24-10772.2002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Sympathetic neurons innervate the heart early in postnatal development, an event that is crucial for proper modulation of blood pressure and cardiac function. However, the axon guidance cues that direct sympathetic neurons to the heart, and the neuronal receptors that recognize those cues, are poorly understood. Here we present evidence that interactions between the alpha4beta1 integrin on sympathetic neurons and vascular cell adhesion molecule-1 (VCAM-1) in the heart plays a role in cardiac innervation. The alpha4 subunit was detected on postnatal rat superior cervical ganglion (SCG) neurons in culture and in cryosections of SCG and heart. VCAM-1 immunoreactivity was detected on cardiac myocytes that associate with invading sympathetic neurons. Purified recombinant soluble VCAM-1 (rsVCAM-1) stimulated SCG neurite outgrowth at levels comparable with laminin 2/4 and fibronectin (Fn), and outgrowth on rs-VCAM-1 and Fn was blocked by antibodies specific for the alpha4 and beta1 integrin subunits. Intrathoracic injection of function-blocking antibodies to alpha4 and VCAM-1, as well as a small molecule inhibitor of alpha4 integrins, significantly reduced sympathetic innervation of the heart. These results indicate that the interaction between alpha4 integrin and VCAM-1 is important for sympathetic innervation of the heart.
Collapse
|
40
|
Pinco KA, He W, Yang JT. alpha4beta1 integrin regulates lamellipodia protrusion via a focal complex/focal adhesion-independent mechanism. Mol Biol Cell 2002; 13:3203-17. [PMID: 12221126 PMCID: PMC124153 DOI: 10.1091/mbc.02-05-0086] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
alpha4beta1 integrin plays an important role in cell migration. We show that when ectopically expressed in Chinese hamster ovary cells, alpha4beta1 is sufficient and required for promoting protrusion of broad lamellipodia in response to scratch-wounding, whereas alpha5beta1 does not have this effect. By time-lapse microscopy of cells expressing an alpha4/green fluorescent protein fusion protein, we show that alpha4beta1 forms transient puncta at the leading edge of cells that begin to protrude lamellipodia in response to scratch-wounding. The cells expressing a mutant alpha4/green fluorescent protein that binds paxillin at a reduced level had a faster response to scratch-wounding, forming alpha4-positive puncta and protruding lamellipodia much earlier. While enhancing lamellipodia protrusion, this mutation reduces random motility of the cells in Transwell assays, indicating that lamellipodia protrusion and random motility are distinct types of motile activities that are differentially regulated by interactions between alpha4beta1 and paxillin. Finally, we show that, at the leading edge, alpha4-positive puncta and paxillin-positive focal complexes/adhesions do not colocalize, but alpha4beta1 and paxillin colocalize partially in ruffles. These findings provide evidence for a specific role of alpha4beta1 in lamellipodia protrusion that is distinct from the motility-promoting functions of alpha5beta1 and other integrins that mediate cell adhesion and signaling events through focal complexes and focal adhesions.
Collapse
Affiliation(s)
- Karen A Pinco
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | |
Collapse
|
41
|
Waitkus-Edwards KR, Martinez-Lemus LA, Wu X, Trzeciakowski JP, Davis MJ, Davis GE, Meininger GA. alpha(4)beta(1) Integrin activation of L-type calcium channels in vascular smooth muscle causes arteriole vasoconstriction. Circ Res 2002; 90:473-80. [PMID: 11884378 DOI: 10.1161/hh0402.105899] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A pathway for the regulation of vascular tone appears to involve coupling between integrins and extracellular matrix proteins or their fragments and the subsequent modulation of ion movement across the smooth muscle cell membrane. Here, we report that the activation of L-type voltage-activated Ca(2+) channels occurs through a novel interaction of alpha(4)beta(1) integrin with peptides containing the Leu-Asp-Val (LDV) integrin--binding sequence, which is found in the CS-1 region of an alternately spliced fibronectin variant. Experiments were conducted on arterioles isolated from rat skeletal muscle. Arterioles exhibited sustained concentration-dependent vasoconstriction to LDV peptides but not to Leu-Glu-Val (LEV) control peptides. The constriction was associated with increased smooth muscle cell [Ca(2+)](i), as measured by using fura 2. The response could be inhibited with a function-blocking anti--alpha(4) integrin antibody. Removal of the endothelium did not alter the vasoconstrictor response. Further experiments demonstrated that the vasoconstriction was abolished by the L-type Ca(2+) channel inhibitor nifedipine and the Src family kinase inhibitor PP2. In studies of isolated smooth muscle cells using whole-cell patch-clamp methods, the L-type current was enhanced by the LDV but not LEV peptide and was blocked by PP2 or antibodies to alpha(4) integrin. Collectively, these data indicate that activation of alpha(4)beta(1) integrin leads to enhanced influx of Ca(2+) through L-type channels by activating a tyrosine kinase pathway, leading to vasoconstriction. Involvement of integrins in the modulation of vascular tone may be particularly important in vascular responses to mechanical signals, such as pressure and flow, and to tissue injury after damage to the extracellular matrix.
Collapse
Affiliation(s)
- Kelli R Waitkus-Edwards
- Department of Medical Physiology, Cardiovascular Research Institute, Division of Vascular Biology, Texas A&M University System Health Science Center, College Station, TX 77843-1114, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Testaz S, Duband JL. Central role of the alpha4beta1 integrin in the coordination of avian truncal neural crest cell adhesion, migration, and survival. Dev Dyn 2001; 222:127-40. [PMID: 11668592 DOI: 10.1002/dvdy.1181] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Based on functional and histological studies, the fibronectin receptor of the integrin family alpha4beta1 has been ascribed a critical role during neural crest cell migration in the vertebrate embryo. In the present study, because integrins have been shown to participate in multiple basic cellular processes, including cell adhesion, migration, survival, proliferation, and differentiation, we have reexamined in detail the role of alpha4beta1 during avian truncal neural crest cell migration. RT-PCR and immunocytochemical studies revealed that migrating neural crest cells but not premigratory cells explanted in vitro expressed detectable levels of alpha4 messengers and proteins suggesting that alpha4beta1 expression was induced at the time of the initiation of the migration phase. In agreement with this observation, antibody inhibition of alpha4beta1 activity in vitro resulted in a strong, immediate and sustained reduction of neural crest cell motion on fibronectin, as judged on videomicroscopy analyses, but apparently did not prevent their delamination from the neural tube. However, alpha4beta1 appeared to exhibit a broader role in the control of cell migration on a variety of extracellular matrix molecules, presumably by regulating cellular events downstream from integrins. Moreover, blocking alpha4beta1 function caused a severe increase in apoptotic cell death among the neural crest population without influencing notably cell proliferation. Collectively, these results indicate that, notwithstanding its critical implication in cell motion, alpha4beta1 integrin could play a central role in neural crest cell development by coordinating multiple cellular events, such as cell adhesion, locomotion, and survival.
Collapse
Affiliation(s)
- S Testaz
- Laboratoire de Biologie du Développement, CNRS UMR 7622, Université Pierre et Marie Curie, 9 quai Saint Bernard, 7éme etage, 75005 Paris, France
| | | |
Collapse
|
43
|
Abstract
The progressive shift from young age to senescence is characterized by structural and functional changes in the cardiac extracellular matrix (ECM), which supports and aligns myocytes and blood vessels, and maintains myocardial mass, structure and function. As cardiac function declines with advancing age, ECM collagen and fibronectin influence diastolic stiffness. ECM binding to membrane-bound receptors, or integrins, directly links ECM to cardiac muscle and fibroblast cells, affording it the permissive role to modulate heart function. To better understand the ECM structure-function relationship in the old heart, we studied the relative protein content of these ECM proteins and integrins across three age groups. Old Balb-c mice (20 months) exhibit biventricular, cardiac hypertrophy, and greater left ventricular (LV) collagen, fibronectin, alpha 1 and alpha 5 integrin protein than middle-aged (12 months) or young (2 months) LV (P<0.05). beta1 integrin protein content is lower in old LV (P<0.05). These data show that advancing age is associated with greater collagen, fibronectin, alpha 1 and alpha 5 integrin content, suggesting that these matrix proteins undergo coordinated regulation in the aging heart. The differential integrin and ECM protein content suggests that there is regulatory signaling to the fibroblasts, which maintain the cardiac ECM.
Collapse
Affiliation(s)
- M L Burgess
- Department of Health Sciences, Laboratory of Cardiovascular Biology, Boston University, 635 Commonwealth Avenue, Boston, MA 02215, USA.
| | | | | |
Collapse
|
44
|
Abstract
We have shown previously that repair in the peripheral nervous system is associated with a reversion to an embryonic pattern of alternative splicing of the extracellular matrix molecule fibronectin. One of the consequent changes is a relative increase in the number of fibronectins expressing the binding site for alpha4 integrins. Here we show that alpha4 integrins are expressed on dorsal root ganglion neuron cell bodies and growth cones in the sciatic nerve during regeneration and that the interaction of alpha4 integrin with alternatively spliced isoforms of recombinant fibronectins containing the alpha4 binding site enhances neurite outgrowth in dorsal root ganglion neurons. The pheochromocytoma (PC12) neuronal cell line, which normally extends neurites poorly on fibronectin, does so efficiently when alpha4 is expressed in the cells. Experiments using chimeric integrins expressed in PC12 cells show that the alpha4 cytoplasmic domain is necessary and sufficient for this enhanced neurite outgrowth. In both dorsal root ganglion neurons and PC12 cells the alpha4 cytoplasmic domain is tightly linked to the intracellular adapter protein paxillin. These experiments suggest an important role for alpha4 integrin and paxillin in peripheral nerve regeneration and show how alternative splicing of fibronectin may provide a mechanism to enhance repair after injury.
Collapse
|
45
|
Muroya S, Nakajima I, Chikuni K. Bovine Skeletal Muscle Cells Predominantly Express a Vascular Cell Adhesion Molecule-1 Seven-Ig Domain Splice Form. Zoolog Sci 2001. [DOI: 10.2108/zsj.18.797] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
46
|
Haack H, Hynes RO. Integrin receptors are required for cell survival and proliferation during development of the peripheral glial lineage. Dev Biol 2001; 233:38-55. [PMID: 11319856 DOI: 10.1006/dbio.2001.0213] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Proliferation and survival of Schwann cells are important for nerve development and for disease processes in peripheral nerves. We have analyzed embryos lacking alpha4- or alpha5-integrins and show here that these integrins contribute to the control of glial cell numbers. To overcome early embryonic lethality an explant and grafting system that allows the study of isolated glial progenitor cells both in vitro and in vivo was used. Schwann cells differentiate in the absence of alpha5 but their numbers and the proliferation rate of early progenitor cells are reduced, suggesting that alpha5 is essential for normal proliferation. Survival, rather than proliferation, is compromised in alpha4-deficient explants. Conditional immortalization allowed further characterization and revealed that alpha4 contributes to survival in a cell-density-dependent fashion. In addition, transplants into chicken embryos were used to analyze in vivo cell migration and showed that cell death occurs mainly in highly motile, individually migrating cells. The cell death patterns in vitro and in vivo argue that alpha4-integrins play a role in survival during cell migration. Neural crest migration has been suggested to require these integrins; however, no defects in migration were observed in the absence of alpha4 or alpha5. We conclude that integrins can complement growth factors in the control of glial cell numbers.
Collapse
Affiliation(s)
- H Haack
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | |
Collapse
|
47
|
Guan K, Czyz J, Fürst DO, Wobus AM. Expression and cellular distribution of alpha(v)integrins in beta(1)integrin-deficient embryonic stem cell-derived cardiac cells. J Mol Cell Cardiol 2001; 33:521-32. [PMID: 11181020 DOI: 10.1006/jmcc.2000.1326] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
beta(1)integrin-deficient (beta(1)-/-) ES cells showed increased differentiation of cardiac cells characterized by reduced adhesion and high beating frequency. Whereas in whole embryoid body outgrowths of beta(1)-/- cells maximum levels of alpha(v), beta(3)and beta(5)integrin mRNA were delayed and transiently upregulated, in cardiac clusters isolated from beta(1)-/- cells, only beta(3)integrin mRNA levels were enhanced in comparison to wild-type (wt) cells. To answer the question, whether alpha(v)and beta(3)integrins may compensate, at least partially, the loss of beta(1)integrin function during cardiac differentiation, the distribution of alpha(v)and beta(3)integrins in beta(1)-/- and wt pacemaker-like cardiac cells was analyzed. A different distribution of alpha(v)and beta(3)integrins in beta(1)-/- v wt cardiac cells was found. In wt cardiac cells, beta(1)integrin was localized in specialized subsarcolemmal regions, in particular, at focal contacts and costameres, but alpha(v)integrin was diffusely distributed. In contrast, in beta(1)-/- cardiac cells, alpha(v)integrin was preponderantly localized at cell membranes, focal contacts and costameres. beta(3)integrin displayed a diffuse pattern both in wt and in beta(1)-/- pacemaker-like cells at early differentiation stages, whereas at terminal stages, beta(3)was colocalized with sarcomeres in wt, but not in beta(1)-/- pacemaker-like cells. Quantitative immunofluorescence analysis revealed increased alpha(v)and beta(3)integrin levels in beta(1)-/- pacemaker-like cardiac cells. Our results led us to conclude that altered cellular distribution of alpha(v)integrin and upregulation of beta(3)integrin correlate with growth and survival of beta(1)-/- cardiac pacemaker-like cells at an early developmental state. However, alpha(v)and beta(3)integrins cannot functionally compensate the loss of beta(1)integrin during terminal differentiation of cardiac cells implicating that cardiomyocytes require specific beta(1)integrin functions for cardiac specialization.
Collapse
Affiliation(s)
- K Guan
- In Vitro Differentiation Group, IPK Gatersleben, D-06466 Gatersleben, Germany
| | | | | | | |
Collapse
|
48
|
Clegg DO, Mullick LH, Wingerd KL, Lin H, Atienza JW, Bradshaw AD, Gervin DB, Cann GM. Adhesive events in retinal development and function: the role of integrin receptors. Results Probl Cell Differ 2001; 31:141-56. [PMID: 10929405 DOI: 10.1007/978-3-540-46826-4_8] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Cells in the developing retina contact a vast array of molecular cues in their microenvironment that are thought to guide their development. Many of these cues are embedded in the surface of neighboring cells or deposited within the extracellular matrix (ECM). Evidence has accumulated that cell-cell and cell-ECM interactions are essential in many phases of neural development, including neuroblast migration, determination of cell fate, axon outgrowth and synapse formation. In this chapter, we examine the developmental and functional roles fulfilled by integrins, a family of receptors for ECM molecules and cell adhesion molecules (CAMs). We have approached this problem by addressing a series of three questions: (1) which integrins are expressed in developing retina? (2) when and where are they expressed? and, (3) what functions do they carry out? Integrins have previously been implicated in axon extension, but new evidence suggests that they are also involved in earlier developmental events in preaxonal neuroblasts. High levels of expression of at least eight integrin subunits have been documented in these young retinal cells, and integrins containing the beta 1 subunit have been implicated in migration of adolescent retinal ganglion cells. Integrin expression persists through adulthood, both in the retina and in the neighboring layer of the retinal pigment epithelium (RPE). The integrin alpha v beta 5 has been shown to reside on the apical surface of the RPE and has been implicated in the phagocytosis of shed photoreceptor outer segments.
Collapse
Affiliation(s)
- D O Clegg
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara 93106, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Ogawa M, Fraser S, Fujimoto T, Endoh M, Nishikawa S, Nishikawa SI. Origin of hematopoietic progenitors during embryogenesis. Int Rev Immunol 2001; 20:21-44. [PMID: 11342296 DOI: 10.3109/08830180109056721] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
It has been widely accepted that hematopoietic and endothelial cell lineages diverge from a common progenitor referred to as the hemangioblast. Recently, analyses of the potential of progenitor cells purified from mouse embryos as well as embryonic stem cells differentiating in vitro resolved intermediate stages between mesodermal cells and committed precursors for hematopoietic and endothelial cell lineages. There are two distinct hematopoietic cell lineages which have different origins, i.e., primitive hematopoietic lineage derived from mesoderm or hemangioblasts and definitive hematopoietic lineage derived from endothelial cells. The endothelium is suggested to provide a milieu in which the definitive hematopoietic lineage acquires multiple potentials.
Collapse
MESH Headings
- Animals
- Antigens, CD/physiology
- Antigens, CD34/metabolism
- Antigens, Differentiation/metabolism
- Biomarkers
- Cadherins/physiology
- Cell Differentiation
- Cell Lineage
- Cells, Cultured
- Core Binding Factor Alpha 2 Subunit
- DNA-Binding Proteins/physiology
- Endothelium, Vascular/cytology
- Fetal Blood/cytology
- Gene Expression Regulation, Developmental
- Hematopoiesis
- Hematopoietic Stem Cells/cytology
- Hematopoietic System/cytology
- Hematopoietic System/embryology
- Humans
- Integrin alpha4
- Mesoderm/cytology
- Mice
- Mice, Knockout
- Models, Biological
- Organ Specificity
- Platelet Endothelial Cell Adhesion Molecule-1/metabolism
- Proto-Oncogene Proteins
- Receptor Protein-Tyrosine Kinases/metabolism
- Receptors, Growth Factor/metabolism
- Receptors, Vascular Endothelial Growth Factor
- Transcription Factors/physiology
- Yolk Sac/cytology
Collapse
Affiliation(s)
- M Ogawa
- Department of Molecular Genetics, Graduate School of Medicine, Kyoto University, Shogoin-Kawaharacho 53, Sakyo-ku, Kyoto 606-8507, Japan.
| | | | | | | | | | | |
Collapse
|
50
|
Pinco KA, Liu S, Yang JT. alpha4 integrin is expressed in a subset of cranial neural crest cells and in epicardial progenitor cells during early mouse development. Mech Dev 2001; 100:99-103. [PMID: 11118892 DOI: 10.1016/s0925-4773(00)00503-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
By RNA in situ hybridization and immunohistochemical analyses of early stage mouse embryos, we find that alpha 4 integrin gene is expressed in migratory cranial neural crest cells originating from the presumptive forebrain, midbrain, and rhombomeres 1 and 2 of the presumptive hindbrain. alpha 4 is also expressed in epicardial progenitor cells in the septum transversum that migrate to the heart.
Collapse
Affiliation(s)
- K A Pinco
- Department of Cell Biology and Anatomy, The Johns Hopkins University School of Medicine, baltimore, MD 21205, USA
| | | | | |
Collapse
|