Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Marion A. Cooley is active.

Publication


Featured researches published by Marion A. Cooley.


EMBO Reports | 2003

Fibulins: physiological and disease perspectives

W. Scott Argraves; Lisa M. Greene; Marion A. Cooley; William M. Gallagher

The fibulins are a family of proteins that are associated with basement membranes and elastic extracellular matrix fibres. This review summarizes findings from studies of animal models of fibulin deficiency, human fibulin gene mutations, human tumours and injury models that have advanced our understanding of the normal and pathological roles of members of this formerly obscure family.


Developmental Cell | 2009

ADAMTS metalloproteases generate active versican fragments that regulate interdigital web regression

Daniel R. McCulloch; Courtney M. Nelson; Laura J. Dixon; Debra L. Silver; James D. Wylie; Volkhard Lindner; Takako Sasaki; Marion A. Cooley; W. Scott Argraves; Suneel S. Apte

We show that combinatorial mouse alleles for the secreted metalloproteases Adamts5, Adamts20 (bt), and Adamts9 result in fully penetrant soft-tissue syndactyly. Interdigital webs in Adamts5(-/-);bt/bt mice had reduced apoptosis and decreased cleavage of the proteoglycan versican; however, the BMP-FGF axis, which regulates interdigital apoptosis was unaffected. BMP4 induced apoptosis, but without concomitant versican proteolysis. Haploinsufficiency of either Vcan or Fbln1, a cofactor for versican processing by ADAMTS5, led to highly penetrant syndactyly in bt mice, suggesting that cleaved versican was essential for web regression. The local application of an aminoterminal versican fragment corresponding to ADAMTS-processed versican, induced cell death in Adamts5(-/-);bt/bt webs. Thus, ADAMTS proteases cooperatively maintain versican proteolysis above a required threshold to create a permissive environment for apoptosis. The data highlight the developmental significance of proteolytic action on the ECM, not only as a clearance mechanism, but also as a means to generate bioactive versican fragments.


Developmental Biology | 2008

Fibulin-1 is required for morphogenesis of neural crest-derived structures

Marion A. Cooley; Christine B. Kern; Victor M. Fresco; Andy Wessels; Robert P. Thompson; Tim C. McQuinn; Waleed O. Twal; Corey H. Mjaatvedt; Christopher J. Drake; W. Scott Argraves

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


BMC Developmental Biology | 2006

Targeted disruption of cubilin reveals essential developmental roles in the structure and function of endoderm and in somite formation

Brian T. Smith; Jason C Mussell; Paul A. Fleming; Jeremy L. Barth; Demetri D. Spyropoulos; Marion A. Cooley; Christopher J. Drake; W. Scott Argraves

BackgroundCubilin is a peripheral membrane protein that interacts with the integral membrane proteins megalin and amnionless to mediate ligand endocytosis by absorptive epithelia such as the extraembryonic visceral endoderm (VE).ResultsHere we report the effects of the genetic deletion of cubilin on mouse embryonic development. Cubilin gene deletion is homozygous embryonic lethal with death occurring between 7.5–13.5 days post coitum (dpc). Cubilin-deficient embryos display developmental retardation and do not advance morphologically beyond the gross appearance of wild-type 8–8.5 dpc embryos. While mesodermal structures such as the allantois and the heart are formed in cubilin mutants, other mesoderm-derived tissues are anomalous or absent. Yolk sac blood islands are formed in cubilin mutants but are unusually large, and the yolk sac blood vessels fail to undergo remodeling. Furthermore, somite formation does not occur in cubilin mutants. Morphological abnormalities of endoderm occur in cubilin mutants and include a stratified epithelium in place of the normally simple columnar VE epithelium and a stratified cuboidal epithelium in place of the normally simple squamous epithelium of the definitive endoderm. Cubilin-deficient VE is also functionally defective, unable to mediate uptake of maternally derived high-density lipoprotein (HDL).ConclusionIn summary, cubilin is required for embryonic development and is essential for the formation of somites, definitive endoderm and VE and for the absorptive function of VE including the process of maternal-embryo transport of HDL.


Journal of The American Society of Nephrology | 2014

Cubilin Maintains Blood Levels of HDL and Albumin

Obaidullah Aseem; Brian T. Smith; Marion A. Cooley; Brent A. Wilkerson; Kelley M. Argraves; Alan T. Remaley; W. Scott Argraves

Cubilin is an endocytic receptor highly expressed in renal proximal tubules, where it mediates uptake of albumin and filtered forms of apoA-I/HDL. Cubilin deficiency leads to urinary loss of albumin and apoA-I; however, the consequences of cubilin loss on the homeostasis of blood albumin and apoA-I/HDL have not been studied. Using mice heterozygous for cubilin gene deletion (cubilin HT mice), we show that cubilin haploinsufficiency leads to reduced renal proximal tubular uptake of albumin and apoA-I and significantly increased urinary loss of albumin and apoA-I. Moreover, cubilin HT mice displayed significantly decreased blood levels of albumin, apoA-I, and HDL. The levels of albumin and apoA-I protein or mRNA expressed in the liver, kidney, or intestine of cubilin HT mice did not change significantly. The clearance rate of small HDL3 particles (density>1.13 g/ml) from the blood increased significantly in cubilin HT mice. In contrast, the rate of clearance of larger HDL2 particles from the blood did not change significantly, indicating a decreased half-life for HDL particles capable of filtering through the glomerulus. On the basis of these findings, we conclude that cubilin deficiency reduces renal salvage and delivery back to the blood of albumin and apoA-I, which decreases blood levels of albumin and apoA-I/HDL. These findings raise the possibility that therapeutic increase of renal cubilin expression might reduce proteinuria and increase blood levels of albumin and HDL.


Developmental Dynamics | 2012

Fibulin-1 is required during cardiac ventricular morphogenesis for versican cleavage, suppression of ErbB2 and Erk1/2 activation and to attenuate trabecular cardiomyocyte proliferation

Marion A. Cooley; Victor M. Fresco; Margaret E. Dorlon; Waleed O. Twal; Nathan V. Lee; Jeremy L. Barth; Christine B. Kern; M. Luisa Iruela-Arispe; W. Scott Argraves

Background: Trabeculation is an integral component of cardiac ventricular morphogenesis and is dependent on the matrix metalloproteinase, ADAMTS1. A substrate of ADAMTS1 is the proteoglycan versican which is expressed in the developing ventricle and which has been implicated in trabeculation. Fibulin‐1 is a versican and ADAMTS1‐binding extracellular matrix protein required for ventricular morphogenesis. Here we investigated the involvement of fibulin‐1 in ADAMTS1‐mediated cleavage of versican in vitro, and the involvement of fibulin‐1 in versican cleavage in ventricular morphogenesis. Results: We show that fibulin‐1 is a cofactor for ADAMTS1‐dependent in vitro cleavage of versican V1, yielding a 70‐kDa amino‐terminal fragment. Furthermore, fibulin‐1‐deficiency in mice was found to cause a significant reduction (>90%) in ventricular levels of the 70‐kDa versican V1 cleavage product and a 2‐fold increase in trabecular cardiomyocyte proliferation. Decreased versican V1 cleavage and augmented trabecular cardiomyocyte proliferation in fibulin‐1 null hearts is accompanied by increased ventricular activation of ErbB2 and Erk1/2. By contrast, versican deficiency was found to lead to decreased cardiomyocyte proliferation and reduced ventricular trabeculation. Conclusion: We conclude that fibulin‐1 regulates versican‐dependent events in ventricular morphogenesis by promoting ADAMTS1 cleavage of versican leading to suppression of trabecular cardiomyocyte proliferation mediated by the ErbB2‐Map kinase pathway. Developmental Dynamics 241:303–314, 2012.


Annals of Biomedical Engineering | 2014

Cellularized microcarriers as adhesive building blocks for fabrication of tubular tissue constructs.

Waleed O. Twal; Sandra C. Klatt; Keerthi Harikrishnan; Ebtesam Gerges; Marion A. Cooley; Thomas C. Trusk; Boran Zhou; Mohamed Gabr; Tarek Shazly; Susan M. Lessner; Roger R. Markwald; W. Scott Argraves

To meet demands of vascular reconstruction, there is a need for prosthetic alternatives to natural blood vessels. Here we explored a new conduit fabrication approach. Macroporous, gelatin microcarriers laden with human umbilical vein endothelial cells and aortic smooth muscle cells were dispensed into tubular agarose molds and found to adhere to form living tubular tissues. The ability of cellularized microcarriers to adhere to one another involved cellular and extracellular matrix bridging that included the formation of epithelium-like cell layers lining the lumenal and ablumenal surfaces of the constructs and the deposition of collagen and elastin fibers. The tubular tissues behaved as elastic solids, with a uniaxial mechanical response that is qualitatively similar to that of native vascular tissues and consistent with their elastin and collagen composition. Linearized measures of the mechanical response of the fabricated tubular tissues at both low and high strains were observed to increase with duration of static culture, with no significant loss of stiffness following decellularization. The findings highlight the utility of cellularized macroporous gelatin microcarriers as self-adhering building blocks for the fabrication of living tubular structures.


Bone | 2014

Fibulin-1 is required for bone formation and Bmp-2-mediated induction of Osterix

Marion A. Cooley; Keerthi Harikrishnan; James A. Oppel; Sloan F. Miler; Jeremy L. Barth; Courtney J. Haycraft; Sakamuri V. Reddy; W. Scott Argraves

The extracellular matrix protein Fibulin-1 (Fbln1) has been shown to be involved in numerous processes including cardiovascular and lung development. Here we have examined the role of Fbln1 in bone formation. Alizarin red staining of skulls from Fbln1-deficient mice showed reduced mineralization of both membranous and endochondral bones. MicroCT (μCT) analysis of the calvarial bones (i.e., frontal, parietal and interparietal bones collectively) indicated that bone volume in Fbln1 nulls at neonatal stage P0 were reduced by 22% (p=0.015). Similarly, Fbln1 null frontal bones showed a 16% (p=0.035) decrease in bone volume, with a reduction in the interfrontal bone, and a discontinuity in the leading edge of the frontal bone. To determine whether Fbln1 played a role in osteoblast differentiation during bone formation, qPCR was used to measure the effects of Fbln1 deficiency on the expression of Osterix (Osx), a transcription factor essential for osteoblast differentiation. This analysis demonstrated that Osx mRNA was significantly reduced in Fbln1-deficient calvarial bones at developmental stages E16.5 (p=0.049) and E17.5 (p=0.022). Furthermore, the ability of Bmp-2 to induce Osx expression was significantly diminished in Fbln1-deficient mouse embryo fibroblasts. Together, these findings indicate that Fbln1 is a new positive modulator of the formation of membranous bone and endochondral bone in the skull, acting as a positive regulator of Bmp signaling.


Mechanisms of Development | 2015

Fibulin-1 suppresses endothelial to mesenchymal transition in the proximal outflow tract

Keerthi Harikrishnan; Marion A. Cooley; Yukiko Sugi; Jeremy L. Barth; Lars Melholt Rasmussen; Christine B. Kern; Kelley M. Argraves; W. Scott Argraves

Endothelial to mesenchymal transition (EMT) that occurs during cardiac outflow tract (OFT) development is critical for formation of the semilunar valves. Fibulin-1 (Fbln1) is an extracellular matrix protein that is present at several sites of EMT, including the OFT (i.e., E9.5-10.5). The aim of this study was to determine the role of Fbln1 in EMT during the earliest events of OFT development. Examination of proximal OFT cushions in Fbln1 null embryos detected hypercellularity at both E9.5 (93% increase; p = 0.002) and E10.5 (43% increase; p = 0.01) as compared to wild type, suggesting that Fbln1 normally suppresses OFT endocardial cushion EMT. This was supported by studies of proximal OFT cushion explants, which showed that explants from Fbln1 null embryos displayed a 58% increase in cells migrating from the explants as compared to wild type (p = 0.005). We next evaluated the effects of Fbln1 deficiency on the expression of factors that regulate proximal OFT EMT. At E9.5, Fbln1 null proximal OFT endocardium and EMT-derived mesenchyme showed increased TGFβ2 (58% increase; p = 0.01) and increased Snail1-positive nuclei (27% increase; p = 0.0003). Histological examination of OFT cushions in Fbln1 null embryos (E9.5) also detected cells present in the cushion that were determined to be erythrocytes based on round morphology, autofluorescence, and positive staining for hemoglobin. Erythrocytes were also detected in Fbln1 null OFT cushions at E10.5. Together, the findings indicate that Fbln1 normally suppresses proximal OFT EMT preventing proximal cushion hypercellularity and blood cell accumulation.


The Journal of Pathology | 2017

Airway remodelling and inflammation in asthma are dependent on the extracellular matrix protein fibulin-1c.

Gang Liu; Marion A. Cooley; Prema M. Nair; Chantal Donovan; Alan C Hsu; Andrew G. Jarnicki; Tatt Jhong Haw; Nicole G. Hansbro; Qi Ge; Alexandra C. Brown; Hock L. Tay; Paul S. Foster; Peter Wark; Jay C. Horvat; Jane E. Bourke; Christopher Grainge; W. Scott Argraves; Brian Oliver; Darryl A. Knight; Janette K. Burgess; Philip M. Hansbro

Asthma is a chronic inflammatory disease of the airways. It is characterized by allergic airway inflammation, airway remodelling, and airway hyperresponsiveness (AHR). Asthma patients, in particular those with chronic or severe asthma, have airway remodelling that is associated with the accumulation of extracellular matrix (ECM) proteins, such as collagens. Fibulin‐1 (Fbln1) is an important ECM protein that stabilizes collagen and other ECM proteins. The level of Fbln1c, one of the four Fbln1 variants, which predominates in both humans and mice, is increased in the serum and airways fluids in asthma but its function is unclear. We show that the level of Fbln1c was increased in the lungs of mice with house dust mite (HDM)‐induced chronic allergic airway disease (AAD). Genetic deletion of Fbln1c and therapeutic inhibition of Fbln1c in mice with chronic AAD reduced airway collagen deposition, and protected against AHR. Fbln1c‐deficient (Fbln1c–/–) mice had reduced mucin (MUC) 5 AC levels, but not MUC5B levels, in the airways as compared with wild‐type (WT) mice. Fbln1c interacted with fibronectin and periostin that was linked to collagen deposition around the small airways. Fbln1c–/– mice with AAD also had reduced numbers of α‐smooth muscle actin‐positive cells around the airways and reduced airway contractility as compared with WT mice. After HDM challenge, these mice also had fewer airway inflammatory cells, reduced interleukin (IL)‐5, IL‐13, IL‐33, tumour necrosis factor (TNF) and CXCL1 levels in the lungs, and reduced IL‐5, IL‐33 and TNF levels in lung‐draining lymph nodes. Therapeutic targeting of Fbln1c reduced the numbers of GATA3‐positive Th2 cells in the lymph nodes and lungs after chronic HDM challenge. Treatment also reduced the secretion of IL‐5 and IL‐13 from co‐cultured dendritic cells and T cells restimulated with HDM extract. Human epithelial cells cultured with Fbln1c peptide produced more CXCL1 mRNA than medium‐treated controls. Our data show that Fbln1c may be a therapeutic target in chronic asthma. Copyright

Collaboration


Dive into the Marion A. Cooley's collaboration.

Top Co-Authors

Avatar

W. Scott Argraves

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Jeremy L. Barth

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Keerthi Harikrishnan

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Waleed O. Twal

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Christine B. Kern

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Victor M. Fresco

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Brian T. Smith

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Christopher J. Drake

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kelley M. Argraves

Medical University of South Carolina

View shared research outputs
Researchain Logo
Decentralizing Knowledge