Network


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

Hotspot


Dive into the research topics where Sharon J. Sequeira is active.

Publication


Featured researches published by Sharon J. Sequeira.


Developmental Biology | 2009

Identification of a mechanochemical checkpoint and negative feedback loop regulating branching morphogenesis.

William P. Daley; Kathryn M. Gulfo; Sharon J. Sequeira; Melinda Larsen

Cleft formation is the initial step in submandibular salivary gland (SMG) branching morphogenesis, and may result from localized actomyosin-mediated cellular contraction. Since ROCK regulates cytoskeletal contraction, we investigated the effects of ROCK inhibition on mouse SMG ex vivo organ cultures. Pharmacological inhibitors of ROCK, isoform-specific ROCK I but not ROCK II siRNAs, as well as inhibitors of myosin II activity stalled clefts at initiation. This finding implies the existence of a mechanochemical checkpoint regulating the transition of initiated clefts into progression-competent clefts. Downstream of the checkpoint, clefts are rendered competent through localized assembly of fibronectin promoted by ROCK I/myosin II. Cleft progression is primarily mediated by ROCK I/myosin II-stimulated cell proliferation with a contribution from cellular contraction. Furthermore, we demonstrate that FN assembly itself promotes epithelial proliferation and cleft progression in a ROCK-dependent manner. ROCK also stimulates a proliferation-independent negative feedback loop to prevent further cleft initiations. These results reveal that cleft initiation and progression are two physically and biochemically distinct processes.


PLOS ONE | 2007

Inhibition of Proliferation by PERK Regulates Mammary Acinar Morphogenesis and Tumor Formation

Sharon J. Sequeira; Aparna C. Ranganathan; Alejandro P. Adam; Bibiana V. Iglesias; Eduardo F. Farias; Julio A. Aguirre-Ghiso

Endoplasmic reticulum (ER) stress signaling can be mediated by the ER kinase PERK, which phosphorylates its substrate eIF2α. This in turn, results in translational repression and the activation of downstream programs that can limit cell growth through cell cycle arrest and/or apoptosis. These responses can also be initiated by perturbations in cell adhesion. Thus, we hypothesized that adhesion-dependent regulation of PERK signaling might determine cell fate. We tested this hypothesis in a model of mammary acini development, a morphogenetic process regulated in part by adhesion signaling. Here we report a novel role for PERK in limiting MCF10A mammary epithelial cell proliferation during acinar morphogenesis in 3D Matrigel culture as well as in preventing mammary tumor formation in vivo. We show that loss of adhesion to a suitable substratum induces PERK-dependent phosphorylation of eIF2α and selective upregulation of ATF4 and GADD153. Further, inhibition of endogenous PERK signaling during acinar morphogenesis, using two dominant-negative PERK mutants (PERK-ΔC or PERK-K618A), does not affect apoptosis but results instead in hyper-proliferative and enlarged lumen-filled acini, devoid of proper architecture. This phenotype correlated with an adhesion-dependent increase in translation initiation, Ki67 staining and upregulation of Laminin-5, ErbB1 and ErbB2 expression. More importantly, the MCF10A cells expressing PERKΔC, but not a vector control, were tumorigenic in vivo upon orthotopic implantation in denuded mouse mammary fat pads. Our results reveal that the PERK pathway is responsive to adhesion-regulated signals and that it is essential for proper acinar morphogenesis and in preventing mammary tumor formation. The possibility that deficiencies in PERK signaling could lead to hyperproliferation of the mammary epithelium and increase the likelihood of tumor formation, is of significance to the understanding of breast cancer.


Biomaterials | 2012

The regulation of focal adhesion complex formation and salivary gland epithelial cell organization by nanofibrous PLGA scaffolds

Sharon J. Sequeira; David A. Soscia; Basak Oztan; Aaron P. Mosier; Riffard Jean-Gilles; Anand Gadre; Nathaniel C. Cady; Bülent Yener; James Castracane; Melinda Larsen

Nanofiber scaffolds have been useful for engineering tissues derived from mesenchymal cells, but few studies have investigated their applicability for epithelial cell-derived tissues. In this study, we generated nanofiber (250 nm) or microfiber (1200 nm) scaffolds via electrospinning from the polymer, poly-l-lactic-co-glycolic acid (PLGA). Cell-scaffold contacts were visualized using fluorescent immunocytochemistry and laser scanning confocal microscopy. Focal adhesion (FA) proteins, such as phosphorylated FAK (Tyr397), paxillin (Tyr118), talin and vinculin were localized to FA complexes in adult cells grown on planar surfaces but were reduced and diffusely localized in cells grown on nanofiber surfaces, similar to the pattern observed in adult mouse salivary gland tissues. Significant differences in epithelial cell morphology and cell clustering were also observed and quantified, using image segmentation and computational cell-graph analyses. No statistically significant differences in scaffold stiffness between planar PLGA film controls compared to nanofibers scaffolds were detected using nanoindentation with atomic force microscopy, indicating that scaffold topography rather than mechanical properties accounts for changes in cell attachments and cell structure. Finally, PLGA nanofiber scaffolds could support the spontaneous self-organization and branching of dissociated embryonic salivary gland cells. Nanofiber scaffolds may therefore have applicability in the future for engineering an artificial salivary gland.


Biology Open | 2013

Quantitative single cell analysis of cell population dynamics during submandibular salivary gland development and differentiation

Deirdre A. Nelson; Charles Manhardt; Vidya Pundalik Kamath; Yunxia Sui; Alberto Santamaria-Pang; Ali Can; Musodiq O. Bello; Alex David Corwin; Sean Richard Dinn; Michael Lazare; Elise M. Gervais; Sharon J. Sequeira; Sarah B. Peters; Fiona Ginty; Michael J. Gerdes; Melinda Larsen

Summary Epithelial organ morphogenesis involves reciprocal interactions between epithelial and mesenchymal cell types to balance progenitor cell retention and expansion with cell differentiation for evolution of tissue architecture. Underlying submandibular salivary gland branching morphogenesis is the regulated proliferation and differentiation of perhaps several progenitor cell populations, which have not been characterized throughout development, and yet are critical for understanding organ development, regeneration, and disease. Here we applied a serial multiplexed fluorescent immunohistochemistry technology to map the progressive refinement of the epithelial and mesenchymal cell populations throughout development from embryonic day 14 through postnatal day 20. Using computational single cell analysis methods, we simultaneously mapped the evolving temporal and spatial location of epithelial cells expressing subsets of differentiation and progenitor markers throughout salivary gland development. We mapped epithelial cell differentiation markers, including aquaporin 5, PSP, SABPA, and mucin 10 (acinar cells); cytokeratin 7 (ductal cells); and smooth muscle &agr;-actin (myoepithelial cells) and epithelial progenitor cell markers, cytokeratin 5 and c-kit. We used pairwise correlation and visual mapping of the cells in multiplexed images to quantify the number of single- and double-positive cells expressing these differentiation and progenitor markers at each developmental stage. We identified smooth muscle &agr;-actin as a putative early myoepithelial progenitor marker that is expressed in cytokeratin 5-negative cells. Additionally, our results reveal dynamic expansion and redistributions of c-kit- and K5-positive progenitor cell populations throughout development and in postnatal glands. The data suggest that there are temporally and spatially discreet progenitor populations that contribute to salivary gland development and homeostasis.


Biomaterials | 2012

Selective functionalization of nanofiber scaffolds to regulate salivary gland epithelial cell proliferation and polarity.

Shraddha I. Cantara; David A. Soscia; Sharon J. Sequeira; Riffard Jean-Gilles; James Castracane; Melinda Larsen

Epithelial cell types typically lose apicobasal polarity when cultured on 2D substrates, but apicobasal polarity is required for directional secretion by secretory cells, such as salivary gland acinar cells. We cultured salivary gland epithelial cells on poly(lactic-co-glycolic acid) (PLGA) nanofiber scaffolds that mimic the basement membrane, a specialized extracellular matrix, and examined cell proliferation and apicobasal polarization. Although cells proliferated on nanofibers, chitosan-coated nanofiber scaffolds stimulated proliferation of salivary gland epithelial cells. Although apicobasal cell polarity was promoted by the nanofiber scaffolds relative to flat surfaces, as determined by the apical localization of ZO-1, it was antagonized by the presence of chitosan. Neither salivary gland acinar nor ductal cells fully polarized on the nanofiber scaffolds, as determined by the homogenous membrane distribution of the mature tight junction marker, occludin. However, nanofiber scaffolds chemically functionalized with the basement membrane protein, laminin-111, promoted more mature tight junctions, as determined by apical localization of occludin, but did not affect cell proliferation. To emulate the multifunctional capabilities of the basement membrane, bifunctional PLGA nanofibers were generated. Both acinar and ductal cell lines responded to signals provided by bifunctional scaffolds coupled to chitosan and laminin-111, demonstrating the applicability of such scaffolds for epithelial cell types.


Journal of Nanotechnology in Engineering and Medicine | 2010

Novel Modeling Approach to Generate a Polymeric Nanofiber Scaffold for Salivary Gland Cells

Riffard Jean-Gilles; David A. Soscia; Sharon J. Sequeira; Michael Melfi; Anand Gadre; James Castracane; Melinda Larsen

BACKGROUND: Electrospun nanofibers have been utilized in many biomedical applications as biomimetics of extracellular matrix proteins that promote self-organization of cells into 3D tissue constructs. As progress towards an artificial salivary gland tissue construct, we prepared nanofiber scaffolds using PLGA, a biodegradable and biocompatible material. METHOD OF APPROACH: We used electrospinning to prepare nanofiber scaffolds using PLGA with both DMF and HFIP as solvents. Using a design of experiment (DOE) approach, system and process parameters were optimized concurrently and their effects on the diameter of the resulting fibers were computed into a single model. A transfer function was used to reproducibly produce nanofibers of a defined diameter, which was confirmed by SEM. The mouse salivary gland epithelial cell line, SIMS, was seeded on the nanofiber scaffolds, and morphology, cell proliferation, and viability were assayed. RESULTS: Varying two or more parameters simultaneously yielded trends diverging from the linear response predicted by previous studies. Comparison of two solvents revealed that the diameter of PLGA nanofibers generated using HFIP is less sensitive to changes in the system and process parameters than are fibers generated using DMF. Inclusion of NaCl reduced morphological inconsistencies and minimized process variability. The resulting nanofiber scaffolds supported attachment, survival and cell proliferation of a mouse salivary gland epithelial cell line. In comparison with glass and flat PLGA films, the nanofibers promoted self-organization of the salivary gland cells into 3D cell clusters, or aggregates. CONCLUSIONS: These data indicate that nanofiber scaffolds promote salivary gland cell organization, and suggest that a nanofiber scaffold could provide a platform for engineering of an artificial salivary gland tissue construct. This study additionally provides a method for efficient production of nanofiber scaffolds for general application in tissue engineering.


Journal of Visualized Experiments | 2013

Genetic Modification and Recombination of Salivary Gland Organ Cultures

Sharon J. Sequeira; Elise M. Gervais; Shayoni Ray; Melinda Larsen

Branching morphogenesis occurs during the development of many organs, and the embryonic mouse submandibular gland (SMG) is a classical model for the study of branching morphogenesis. In the developing SMG, this process involves iterative steps of epithelial bud and duct formation, to ultimately give rise to a complex branched network of acini and ducts, which serve to produce and modify/transport the saliva, respectively, into the oral cavity. The epithelial-associated basement membrane and aspects of the mesenchymal compartment, including the mesenchyme cells, growth factors and the extracellular matrix, produced by these cells, are critical to the branching mechanism, although how the cellular and molecular events are coordinated remains poorly understood. The study of the molecular mechanisms driving epithelial morphogenesis advances our understanding of developmental mechanisms and provides insight into possible regenerative medicine approaches. Such studies have been hampered due to the lack of effective methods for genetic manipulation of the salivary epithelium. Currently, adenoviral transduction represents the most effective method for targeting epithelial cells in adult glands in vivo. However, in embryonic explants, dense mesenchyme and the basement membrane surrounding the epithelial cells impedes viral access to the epithelial cells. If the mesenchyme is removed, the epithelium can be transfected using adenoviruses, and epithelial rudiments can resume branching morphogenesis in the presence of Matrigel or laminin-111. Mesenchyme-free epithelial rudiment growth also requires additional supplementation with soluble growth factors and does not fully recapitulate branching morphogenesis as it occurs in intact glands. Here we describe a technique which facilitates adenoviral transduction of epithelial cells and culture of the transfected epithelium with associated mesenchyme. Following microdissection of the embryonic SMGs, removal of the mesenchyme, and viral infection of the epithelium with a GFP-containing adenovirus, we show that the epithelium spontaneously recombines with uninfected mesenchyme, recapitulating intact SMG glandular structure and branching morphogenesis. The genetically modified epithelial cell population can be easily monitored using standard fluorescence microscopy methods, if fluorescently-tagged adenoviral constructs are used. The tissue recombination method described here is currently the most effective and accessible method for transfection of epithelial cells with a wild-type or mutant vector within a complex 3D tissue construct that does not require generation of transgenic animals.


BMC Cell Biology | 2009

Inhibition of eIF2α dephosphorylation inhibits ErbB2-induced deregulation of mammary acinar morphogenesis

Sharon J. Sequeira; Huei Chi Wen; Alvaro Avivar-Valderas; Eduardo F. Farias; Julio A. Aguirre-Ghiso

BackgroundThe ErbB2/Her2/Neu receptor tyrosine kinase is amplified in ~30% of human breast cancers. Phosphorylation of the translation initiation factor, eIF2α inhibits global protein synthesis and activates a stress signaling and growth suppressive program. We have shown that forced phosphorylation of eIF2α can suppress head and neck, colorectal carcinoma and multiple myeloma tumor growth and/or survival. Here we explore whether ErbB2 modulates eIF2α phosphorylation and whether forced phosphorylation of the latter can antagonize ErbB2 deregulation of mammary acinar morphogenesis.ResultsWe tested whether ErbB2 signaling influenced eIF2α signaling and whether enhanced phosphorylation of the latter affected ErbB2-deregulated mammary acinar development. We obtained stable MCF10A cells overexpressing wild-type (Wt) Neu/ErbB2 or a constitutively active (CA) variant via retroviral delivery or mammary tumor cells from MMTV-Neu tumors. Western blotting, RT-PCR and confocal microscopy were used to analyze the effects of ErbB2 activation on eIF2α signaling and the effect of the GADD34-PP1C inhibitor salubrinal. Wt- and MMTV-Neu cells formed aberrant acini structures resembling DCIS, while CA-ErbB2 overexpression induced invasive lesions. In these structures we found that CA-ErbB2 but not the Wt variant significantly down-regulated the pro-apoptotic gene CHOP. This occurred without apparent modulation of basal phosphorylation of PERK and eIF2α or induction of its downstream target ATF4. However, inhibition of eIF2α dephosphorylation with salubrinal was sufficient to inhibit Wt- and CA-ErbB2- as well as MMTV-Neu-induced deregulation of acinar growth. This was linked to enhanced CHOP expression, inhibition of proliferation, induction of apoptosis and luminal clearing in Wt-ErbB2 and to inhibition of cyclin D1 levels and subsequent proliferation in CA-ErbB2 cells.ConclusionDepending on the strength of ErbB2 signaling there is a differential regulation of CHOP and eIF2α phosphorylation. ErbB2 uncouples in basal conditions eIF2α phosphorylation from CHOP induction. However, this signal was restored by salubrinal treatment in Wt-ErbB2 expressing MCF10A cells as these DCIS-like structures underwent luminal clearing. In CA-ErbB2 structures apoptosis is not induced by salubrinal and instead a state of quiescence with reduced proliferation was achieved. Treatments that stabilize P-eIF2α levels may be effective in treating ErbB2 positive cancers without severely disrupting normal tissue function and structure.


Journal of Cell Science | 2014

Integrins promote cytokinesis through the RSK signaling axis

Shomita S. Mathew; Bethsaida Nieves; Sharon J. Sequeira; Savitha Sambandamoorthy; Kevin Pumiglia; Melinda Larsen; Susan E. LaFlamme

ABSTRACT Cytokinesis is the final stage in cell division. Although integrins can regulate cytokinesis, the mechanisms involved are not fully understood. In this study, we demonstrate that integrin-regulated ERK (extracellular signal-related kinase) and RSK (p90 ribosomal S6 kinase) signaling promotes successful cytokinesis. Inhibiting the activation of ERK and RSK in CHO cells by a mutation in the integrin &bgr;1 cytoplasmic tail or with pharmacological inhibitors results in the accumulation of cells with midbodies and the formation of binucleated cells. Activation of ERK and RSK signaling by the expression of constitutively active RAF1 suppresses the mutant phenotype in a RSK-dependent manner. Constitutively active RSK2 also restores cytokinesis inhibited by the mutant integrin. Importantly, the regulatory role of the RSK pathway is not specific to CHO cells. MCF-10A human mammary epithelial cells and HPNE human pancreatic ductal epithelial cells exhibit a similar dependence on RSK for successful cytokinesis. In addition, depriving mitotic MCF10A cells of integrin-mediated adhesion by incubating them in suspension suppressed ERK and RSK activation and resulted in a failure of cytokinesis. Furthermore, inhibition of RSK or integrins within the 3D context of a developing salivary gland organ explant also leads to an accumulation of epithelial cells with midbodies, suggesting a similar defect in cytokinesis. Interestingly, neither ERK nor RSK regulates cytokinesis in human fibroblasts, suggesting cell-type specificity. Taken together, our results identify the integrin–RSK signaling axis as an important regulator of cytokinesis in epithelial cells. We propose that the proper interaction of cells with their microenvironment through integrins contributes to the maintenance of genomic stability by promoting the successful completion of cytokinesis.


international symposium on biomedical imaging | 2010

Cell-graph modeling of salivary gland morphology

Cemal Cagatay Bilgin; Shayoni Ray; William P. Daley; Banu Baydil; Sharon J. Sequeira; Bülent Yener; Melinda Larsen

Branching morphogenesis is a developmental process shared by many organs, including the submandibular salivary gland. During morphogenesis, cells within the gland undergo rearrangements to cause changes in the overall tissue morphology. This work presents a methodology based on cell-graphs to quantify these changes in cellular arrangements. Multiple confocal images of developing salivary gland organ cultures are captured. These cultures are immunostained with a nuclear marker and an epithelial marker to identify epithelial cells as separate from mesenchymal cells. Confocal images are stitched and segmented to identify epithelial and mesenchymal nuclei. Cell-graphs are constructed to model the structural organization of epithelial and mesenchymal cells. Cell-graph metrics are calculated to extract mathematical features that discriminate epithelial vs mesenchymal cells organizations and also distinguish between glands treated with pharmacological inhibitors vs vehicle control. The results indicate that cell-graph features can be used to both describe and predict the developing salivary gland to provide insights into cellular and physical processes driving morphogenesis.

Collaboration


Dive into the Sharon J. Sequeira's collaboration.

Top Co-Authors

Avatar

Melinda Larsen

State University of New York System

View shared research outputs
Top Co-Authors

Avatar

David A. Soscia

State University of New York System

View shared research outputs
Top Co-Authors

Avatar

James Castracane

State University of New York System

View shared research outputs
Top Co-Authors

Avatar

Julio A. Aguirre-Ghiso

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Riffard Jean-Gilles

State University of New York System

View shared research outputs
Top Co-Authors

Avatar

Elise M. Gervais

State University of New York System

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bülent Yener

Rensselaer Polytechnic Institute

View shared research outputs
Top Co-Authors

Avatar

Eduardo F. Farias

Icahn School of Medicine at Mount Sinai

View shared research outputs
Researchain Logo
Decentralizing Knowledge