Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Organogenesis ; 12(4): 194-216, 2016 10.
Article in English | MEDLINE | ID: mdl-27841695

ABSTRACT

The salivary epithelium initiates as a solid mass of epithelial cells that are organized into a primary bud that undergoes morphogenesis and differentiation to yield bilayered acini consisting of interior secretory acinar cells that are surrounded by contractile myoepithelial cells in mature salivary glands. How the primary bud transitions into acini has not been previously documented. We document here that the outer epithelial cells subsequently undergo a vertical compression as they express smooth muscle α-actin and differentiate into myoepithelial cells. The outermost layer of polarized epithelial cells assemble and organize the basal deposition of basement membrane, which requires basal positioning of the polarity protein, Par-1b. Whether Par-1b is required for the vertical compression and differentiation of the myoepithelial cells is unknown. Following manipulation of Par-1b in salivary gland organ explants, Par-1b-inhibited explants showed both a reduced vertical compression of differentiating myoepithelial cells and reduced levels of smooth muscle α-actin. Rac1 knockdown and inhibition of Rac GTPase function also inhibited branching morphogenesis. Since Rac regulates cellular morphology, we investigated a contribution for Rac in myoepithelial cell differentiation. Inhibition of Rac GTPase activity showed a similar reduction in vertical compression and smooth muscle α-actin levels while decreasing the levels of Par-1b protein and altering its basal localization in the outer cells. Inhibition of ROCK, which is required for basal positioning of Par-1b, resulted in mislocalization of Par-1b and loss of vertical cellular compression, but did not significantly alter levels of smooth muscle α-actin in these cells. Overexpression of Par-1b in the presence of Rac inhibition restored basement membrane protein levels and localization. Our results indicate that the basal localization of Par-1b in the outer epithelial cells is required for myoepithelial cell compression, and Par-1b is required for myoepithelial differentiation, regardless of its localization.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Differentiation , Epithelial Cells/cytology , Morphogenesis , Protein Serine-Threonine Kinases/metabolism , Salivary Glands/cytology , Salivary Glands/embryology , Animals , Basement Membrane/metabolism , Cell Polarity , Cell Proliferation , Epithelial Cells/metabolism , Mice , Models, Biological , rac GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism
2.
Biomaterials ; 34(28): 6773-84, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23777914

ABSTRACT

There is a need for an artificial salivary gland as a long-term remedy for patients suffering from salivary hypofunction, a leading cause of chronic xerostomia (dry mouth). Current salivary gland tissue engineering approaches are limited in that they either lack sufficient physical cues and surface area needed to facilitate epithelial cell differentiation, or they fail to provide a mechanism for assembling an interconnected branched network of cells. We have developed highly-ordered arrays of curved hemispherical "craters" in polydimethylsiloxane (PDMS) using wafer-level integrated circuit (IC) fabrication processes, and lined them with electrospun poly-lactic-co-glycolic acid (PLGA) nanofibers, designed to mimic the three-dimensional (3-D) in vivo architecture of the basement membrane surrounding spherical acini of salivary gland epithelial cells. These micropatterned scaffolds provide a method for engineering increased surface area and were additionally investigated for their ability to promote cell polarization. Two immortalized salivary gland cell lines (SIMS, ductal and Par-C10, acinar) were cultured on fibrous crater arrays of various radii and compared with those grown on flat PLGA nanofiber substrates, and in 3-D Matrigel. It was found that by increasing crater curvature, the average height of the cell monolayer of SIMS cells and to a lesser extent, Par-C10 cells, increased to a maximum similar to that seen in cells grown in 3-D Matrigel. Increasing curvature resulted in higher expression levels of tight junction protein occludin in both cell lines, but did not induce a change in expression of adherens junction protein E-cadherin. Additionally, increasing curvature promoted polarity of both cell lines, as a greater apical localization of occludin was seen in cells on substrates of higher curvature. Lastly, substrate curvature increased expression of the water channel protein aquaporin-5 (Aqp-5) in Par-C10 cells, suggesting that curved nanofiber substrates are more suitable for promoting differentiation of salivary gland cells.


Subject(s)
Lactic Acid/chemistry , Nanofibers/chemistry , Polymers/chemistry , Salivary Glands/cytology , Tissue Engineering , Animals , Blotting, Western , Cell Differentiation/physiology , Cells, Cultured , Immunohistochemistry , Mice , Microscopy, Confocal , Microscopy, Electron, Scanning , Polyesters , Rats
3.
Biol Open ; 2(5): 439-47, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23789091

ABSTRACT

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 α-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 α-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.

4.
J Vis Exp ; (71): e50060, 2013 Jan 28.
Article in English | MEDLINE | ID: mdl-23407326

ABSTRACT

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.


Subject(s)
Organ Culture Techniques/methods , Submandibular Gland/physiology , Transfection/methods , Adenoviridae/genetics , Animals , Epithelial Cells/physiology , Female , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Mice , Microdissection/methods , Pregnancy
5.
Biomaterials ; 33(33): 8372-82, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22938763

ABSTRACT

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.


Subject(s)
Epithelial Cells/cytology , Nanofibers/chemistry , Salivary Glands/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Cell Polarity/physiology , Cell Proliferation , Immunoblotting , Immunohistochemistry , Lactic Acid/chemistry , Microscopy, Confocal , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Rats
6.
Biomaterials ; 33(11): 3175-86, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22285464

ABSTRACT

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.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/physiology , Focal Adhesions/physiology , Lactic Acid/chemistry , Nanotubes/chemistry , Polyglycolic Acid/chemistry , Salivary Glands/cytology , Tissue Engineering/instrumentation , Tissue Scaffolds , Animals , Cell Adhesion/physiology , Cell Survival , Cells, Cultured , Mice , Nanotubes/ultrastructure , Polylactic Acid-Polyglycolic Acid Copolymer , Salivary Glands/physiology
7.
Front Oral Biol ; 14: 48-77, 2010.
Article in English | MEDLINE | ID: mdl-20428011

ABSTRACT

The interstitial extracellular matrix (ECM) and epithelial-cell associated basement membrane (BM) play critical roles in the morphogenesis and differentiation of developing salivary glands. Early studies used ex vivo organ culture and tissue recombination methods to identify the importance of the ECM in organ development. Incorporation of transgenic mice and molecular tools has facilitated progress in our understanding of the mechanisms by which ECM proteins influence SMG development. Recent work has identified alterations in the ECM, BM, and associated proteins in salivary gland diseases, including Sjögren's syndrome and salivary gland cancers, but the significance of such changes is not known. Understanding the basic mechanisms controlling morphogenesis and differentiation in mammalian organ development is the first step towards understanding pathogenesis. Molecular characterization of the function of the ECM and BM in cellular processes is critical for future development of therapeutic approaches in regenerative medicine and tissue engineering. Here we provide a historical overview of experiments defining the functions of the ECM, ECM receptors, and associated molecules in salivary gland development. We include a discussion of the function of ECM-associated proteases and major growth factor signaling components that are in some way regulated by the ECM or associated molecules. We conclude with a discussion of defects in ECM and BM occurring in salivary gland pathologies and speculation on future areas of research pertaining to further understanding of the function of the ECM in the salivary gland.


Subject(s)
Extracellular Matrix Proteins/physiology , Intercellular Signaling Peptides and Proteins/physiology , Organogenesis/physiology , Salivary Glands/embryology , Animals , Basement Membrane/physiology , Intracellular Signaling Peptides and Proteins/physiology , Mice , Mice, Transgenic , Morphogenesis/physiology , Salivary Gland Diseases/physiopathology , Salivary Proteins and Peptides/physiology
8.
Dev Biol ; 336(2): 169-82, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19804774

ABSTRACT

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.


Subject(s)
Morphogenesis , Submandibular Gland/embryology , Animals , Apoptosis , Blotting, Western , Cell Line , Cell Proliferation , Immunohistochemistry , Mice , RNA, Small Interfering , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
9.
BMC Cell Biol ; 10: 64, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19754954

ABSTRACT

BACKGROUND: The ErbB2/Her2/Neu receptor tyrosine kinase is amplified in approximately 30% of human breast cancers. Phosphorylation of the translation initiation factor, eIF2alpha inhibits global protein synthesis and activates a stress signaling and growth suppressive program. We have shown that forced phosphorylation of eIF2alpha can suppress head and neck, colorectal carcinoma and multiple myeloma tumor growth and/or survival. Here we explore whether ErbB2 modulates eIF2alpha phosphorylation and whether forced phosphorylation of the latter can antagonize ErbB2 deregulation of mammary acinar morphogenesis. RESULTS: We tested whether ErbB2 signaling influenced eIF2alpha 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 eIF2alpha 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 eIF2alpha or induction of its downstream target ATF4. However, inhibition of eIF2alpha 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. CONCLUSION: Depending on the strength of ErbB2 signaling there is a differential regulation of CHOP and eIF2alpha phosphorylation. ErbB2 uncouples in basal conditions eIF2alpha 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-eIF2alpha levels may be effective in treating ErbB2 positive cancers without severely disrupting normal tissue function and structure.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Mammary Glands, Animal/metabolism , Morphogenesis , Receptor, ErbB-2/metabolism , Signal Transduction , Activating Transcription Factor 4/metabolism , Animals , Apoptosis , Cells, Cultured , Cyclin D1/genetics , Eukaryotic Initiation Factor-2/genetics , Female , Humans , Mammary Glands, Animal/cytology , Mammary Glands, Animal/growth & development , Mice , Phosphorylation , Receptor, ErbB-2/genetics , Transcription Factor CHOP/metabolism
10.
PLoS One ; 2(7): e615, 2007 Jul 18.
Article in English | MEDLINE | ID: mdl-17637831

ABSTRACT

Endoplasmic reticulum (ER) stress signaling can be mediated by the ER kinase PERK, which phosphorylates its substrate eIF2alpha. 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 eIF2alpha and selective upregulation of ATF4 and GADD153. Further, inhibition of endogenous PERK signaling during acinar morphogenesis, using two dominant-negative PERK mutants (PERK-DeltaC 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 PERKDeltaC, 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.


Subject(s)
eIF-2 Kinase/metabolism , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Apoptosis , Breast/enzymology , Cell Cycle , Cell Line , Endoplasmic Reticulum/enzymology , Epithelial Cells/cytology , Epithelial Cells/enzymology , Eukaryotic Initiation Factor-2/metabolism , Female , Gene Expression Regulation , Humans , Mice , Phosphorylation , RNA, Messenger/genetics , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...