101
|
Yang TL, Hsiao YC. Chitosan facilitates structure formation of the salivary gland by regulating the basement membrane components. Biomaterials 2015; 66:29-40. [PMID: 26189212 DOI: 10.1016/j.biomaterials.2015.06.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 06/02/2015] [Accepted: 06/03/2015] [Indexed: 12/16/2022]
Abstract
Tissue structure is important for inherent physiological function and should be recapitulated during tissue engineering for regenerative purposes. The salivary gland is a branched organ that is responsible for saliva secretion and regulation. The salivary glands develop from epithelial-mesenchymal interactions, and depend on the support of the basement membrane (BM). Chitosan-based biomaterials have been demonstrated to be competent in facilitating the formation of salivary gland tissue structure. However, the underlying mechanisms have remained elusive. In the developing submandibular gland (SMG), the chitosan effect was found to diminish when collagen and laminin were removed from cultured SMG explants. Chitosan increased the expression of BM components including collagen, laminin, and heparan sulfate proteoglycan, and also facilitated BM components and the corresponding receptors to be expressed in tissue-specific patterns beneficial for SMG branching. The chitosan effect decreased when either laminin components or receptors were inhibited, as well when the downstream signaling was blocked. Our results revealed that chitosan promotes salivary glands branching through the BM. By regulating BM components and receptors, chitosan efficiently stimulated downstream signaling to facilitate salivary gland branching. The present study revealed the underlying mechanism of the chitosan effect in engineering SMG structure formation.
Collapse
Affiliation(s)
- Tsung-Lin Yang
- Department of Otolaryngology, National Taiwan University Hospital, College of Medicine, Taipei, Taiwan; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan.
| | - Ya-Chuan Hsiao
- Department of Ophthalmology, Zhongxing Branch, Taipei City Hospital, Taipei, Taiwan; Department of Ophthalmology, College of Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
102
|
Knosp WM, Knox SM, Lombaert IMA, Haddox CL, Patel VN, Hoffman MP. Submandibular parasympathetic gangliogenesis requires sprouty-dependent Wnt signals from epithelial progenitors. Dev Cell 2015; 32:667-77. [PMID: 25805134 DOI: 10.1016/j.devcel.2015.01.023] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 11/21/2014] [Accepted: 01/21/2015] [Indexed: 10/23/2022]
Abstract
Parasympathetic innervation is critical for submandibular gland (SMG) development and regeneration. Parasympathetic ganglia (PSG) are derived from Schwann cell precursors that migrate along nerves, differentiate into neurons, and coalesce within their target tissue to form ganglia. However, signals that initiate gangliogenesis after the precursors differentiate into neurons are unknown. We found that deleting negative regulators of FGF signaling, Sprouty1 and Sprouty2 (Spry1/2DKO), resulted in a striking loss of gangliogenesis, innervation, and keratin 5-positive (K5+) epithelial progenitors in the SMG. Here we identify Wnts produced by K5+ progenitors in the SMG as key mediators of gangliogenesis. Wnt signaling increases survival and proliferation of PSG neurons, and inhibiting Wnt signaling disrupts gangliogenesis and organ innervation. Activating Wnt signaling and reducing FGF gene dosage rescues gangliogenesis and innervation in both the Spry1/2DKO SMG and pancreas. Thus, K5+ progenitors produce Wnt signals to establish the PSG-epithelial communication required for organ innervation and progenitor cell maintenance.
Collapse
Affiliation(s)
- Wendy M Knosp
- Matrix and Morphogenesis Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Sarah M Knox
- Matrix and Morphogenesis Section, NIDCR, NIH, Bethesda, MD 20892, USA; Department of Cell and Tissue Biology, UCSF, San Francisco, CA 94143, USA
| | | | - Candace L Haddox
- Matrix and Morphogenesis Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Vaishali N Patel
- Matrix and Morphogenesis Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Matthew P Hoffman
- Matrix and Morphogenesis Section, NIDCR, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
103
|
Dabrowski A, Terauchi A, Strong C, Umemori H. Distinct sets of FGF receptors sculpt excitatory and inhibitory synaptogenesis. Development 2015; 142:1818-30. [PMID: 25926357 PMCID: PMC4440923 DOI: 10.1242/dev.115568] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 03/25/2015] [Indexed: 12/13/2022]
Abstract
Neurons in the brain must establish a balanced network of excitatory and inhibitory synapses during development for the brain to function properly. An imbalance between these synapses underlies various neurological and psychiatric disorders. The formation of excitatory and inhibitory synapses requires precise molecular control. In the hippocampus, the structure crucial for learning and memory, fibroblast growth factor 22 (FGF22) and FGF7 specifically promote excitatory or inhibitory synapse formation, respectively. Knockout of either Fgf gene leads to excitatory-inhibitory imbalance in the mouse hippocampus and manifests in an altered susceptibility to epileptic seizures, underscoring the importance of FGF-dependent synapse formation. However, the receptors and signaling mechanisms by which FGF22 and FGF7 induce excitatory and inhibitory synapse differentiation are unknown. Here, we show that distinct sets of overlapping FGF receptors (FGFRs), FGFR2b and FGFR1b, mediate excitatory or inhibitory presynaptic differentiation in response to FGF22 and FGF7. Excitatory presynaptic differentiation is impaired in Fgfr2b and Fgfr1b mutant mice; however, inhibitory presynaptic defects are only found in Fgfr2b mutants. FGFR2b and FGFR1b are required for an excitatory presynaptic response to FGF22, whereas only FGFR2b is required for an inhibitory presynaptic response to FGF7. We further find that FGFRs are required in the presynaptic neuron to respond to FGF22, and that FRS2 and PI3K, but not PLCγ, mediate FGF22-dependent presynaptic differentiation. Our results reveal the specific receptors and signaling pathways that mediate FGF-dependent presynaptic differentiation, and thereby provide a mechanistic understanding of precise excitatory and inhibitory synapse formation in the mammalian brain.
Collapse
MESH Headings
- Animals
- Cell Differentiation/genetics
- Cell Differentiation/physiology
- Cells, Cultured
- Fibroblast Growth Factors/genetics
- Fibroblast Growth Factors/metabolism
- Mice
- Mice, Knockout
- Neurogenesis/genetics
- Neurogenesis/physiology
- Neurons/cytology
- Neurons/metabolism
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 2/metabolism
- Receptors, Fibroblast Growth Factor/genetics
- Receptors, Fibroblast Growth Factor/metabolism
- Synapses/metabolism
Collapse
Affiliation(s)
- Ania Dabrowski
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA Medical Scientist Training Program, University of Michigan Medical School, Ann Arbor, MI 48109-2200, USA Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109-2200, USA Molecular & Behavioral Neuroscience Institute, University of Michigan Medical School, Ann Arbor, MI 48109-2200, USA
| | - Akiko Terauchi
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA Molecular & Behavioral Neuroscience Institute, University of Michigan Medical School, Ann Arbor, MI 48109-2200, USA
| | - Cameron Strong
- Molecular & Behavioral Neuroscience Institute, University of Michigan Medical School, Ann Arbor, MI 48109-2200, USA
| | - Hisashi Umemori
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA Medical Scientist Training Program, University of Michigan Medical School, Ann Arbor, MI 48109-2200, USA Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109-2200, USA Molecular & Behavioral Neuroscience Institute, University of Michigan Medical School, Ann Arbor, MI 48109-2200, USA Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109-2200, USA
| |
Collapse
|
104
|
Mattingly A, Finley JK, Knox SM. Salivary gland development and disease. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2015; 4:573-90. [PMID: 25970268 DOI: 10.1002/wdev.194] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 04/14/2015] [Accepted: 04/15/2015] [Indexed: 12/21/2022]
Abstract
Mammalian salivary glands synthesize and secrete saliva via a vast interconnected network of epithelial tubes attached to secretory end units. The extensive morphogenesis required to establish this organ is dependent on interactions between multiple cell types (epithelial, mesenchymal, endothelial, and neuronal) and the engagement of a wide range of signaling pathways. Here we describe critical regulators of salivary gland development and discuss how mutations in these impact human organogenesis. In particular, we explore the genetic contribution of growth factor pathways, nerve-derived factors and extracellular matrix molecules to salivary gland formation in mice and humans.
Collapse
Affiliation(s)
- Aaron Mattingly
- Department of Cell & Tissue Biology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jennifer K Finley
- Department of Cell & Tissue Biology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Sarah M Knox
- Department of Cell & Tissue Biology, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
105
|
Ibuka S, Matsumoto S, Fujii S, Kikuchi A. The P2Y₂ receptor promotes Wnt3a- and EGF-induced epithelial tubular formation by IEC6 cells by binding to integrins. J Cell Sci 2015; 128:2156-68. [PMID: 25908848 DOI: 10.1242/jcs.169060] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 04/13/2015] [Indexed: 12/15/2022] Open
Abstract
Epithelial tubular structures are essential units in various organs. Here, we used rat intestinal epithelial IEC6 cells to investigate tubulogenesis and we found that tubular formation was induced by a combination of Wnt3a and EGF signaling during three-dimensional culture. Wnt3a and EGF induced the expression of the P2Y2 receptor (P2Y2R, also known as P2RY2), and knockdown of P2Y2R suppressed tubular formation. A P2Y2R mutant that lacks nucleotide responsiveness rescued the phenotypes resulting from P2Y2R knockdown, suggesting that nucleotide-dependent responses are not required for P2Y2R functions in tubular formation. The Arg-Gly-Asp (RGD) sequence of P2Y2R has been shown to interact with integrins, and a P2Y2R mutant lacking integrin-binding activity was unable to induce tubular formation. P2Y2R expression inhibited the interaction between integrins and fibronectin, and induced cell morphological changes and proliferation. Inhibition of integrin and fibronectin binding by treatment with the cyclic RGD peptide and fibronectin knockdown induced tubular formation in the presence of EGF alone, but a fibronectin coat suppressed Wnt3a- and EGF-induced tubular formation. These results suggest that Wnt3a- and EGF-induced P2Y2R expression causes tubular formation by preventing the binding of integrins and fibronectin rather than by mediating nucleotide responses.
Collapse
Affiliation(s)
- Souji Ibuka
- Departments of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan Pediatric Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Shinji Matsumoto
- Departments of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Shinsuke Fujii
- Departments of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Akira Kikuchi
- Departments of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
106
|
The contribution of specific cell subpopulations to submandibular salivary gland branching morphogenesis. Curr Opin Genet Dev 2015; 32:47-54. [PMID: 25706196 DOI: 10.1016/j.gde.2015.01.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 01/25/2015] [Accepted: 01/27/2015] [Indexed: 11/23/2022]
Abstract
Branching morphogenesis is the developmental program responsible for generating a large surface to volume ratio in many secretory and absorptive organs. To accomplish branching morphogenesis, spatiotemporal regulation of specific cell subpopulations is required. Here, we review recent studies that define the contributions of distinct cell subpopulations to specific cellular processes during branching morphogenesis in the mammalian submandibular salivary gland, including the initiation of the gland, the coordination of cleft formation, and the contribution of stem/progenitor cells to morphogenesis. In conclusion, we provide an overview of technological advances that have opened opportunities to further probe the contributions of specific cell subpopulations and to define the integration of events required for branching morphogenesis.
Collapse
|
107
|
Chung D, Gao F, Jegga AG, Das SK. Estrogen mediated epithelial proliferation in the uterus is directed by stromal Fgf10 and Bmp8a. Mol Cell Endocrinol 2015; 400:48-60. [PMID: 25451979 PMCID: PMC4751583 DOI: 10.1016/j.mce.2014.11.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 11/03/2014] [Accepted: 11/03/2014] [Indexed: 11/25/2022]
Abstract
To define endometrial stromal-derived paracrine mediators that participate in estradiol-17β (E2)-induced epithelial proliferation, microarray analysis of gene expression was carried out in mouse uterine epithelial-stromal co-culture systems under the condition of E2 or vehicle (control). Our results demonstrated gene alteration by E2: in epithelial cells, we found up-regulation of 119 genes and down-regulation of 28 genes, while in stroma cells we found up-regulation of 144 genes and down-regulation of 184 genes. A functional enrichment analysis of the upregulated epithelial genes implicated them for proliferation, while upregulated stromal genes were associated with extracellular functions. Quantitative RT-PCR and in situ hybridization results confirmed differential gene expression in both cell cultures and ovariectomized uteri after the above treatments. Based on our identification of stromal secretory factors, we found evidence that suppression by siRNA specifically for Bmp8a and/or Fgf10 in the stromal layer caused significant inhibition of proliferation by E2 in the co-culture system, suggesting Bmp8a and Fgf10 act as paracrine mediators during E2-dependent control of uterine proliferation. The localization of receptors and receptor activation signaling in epithelial cells in both the co-culture system and uteri was consistent with their involvement in ligand-receptor signaling. Interestingly, loss of Bmp8a or Fgf10 also caused abrogation of E2-regulated epithelial receptor signaling in co-culture systems, suggesting that stroma-derived Fgf10 and Bmp8a are responsible for epithelial communication. Overall, stromal Fgf10 and Bmp8a serve as potential paracrine factors for E2-dependent regulation of epithelial proliferation in the uterus.
Collapse
Affiliation(s)
- Daesuk Chung
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; The Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Fei Gao
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; The Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Anil G Jegga
- Biomedical Informatics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Sanjoy K Das
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; The Perinatal Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA.
| |
Collapse
|
108
|
May AJ, Chatzeli L, Proctor GB, Tucker AS. Salivary Gland Dysplasia in Fgf10 Heterozygous Mice: A New Mouse Model of Xerostomia. Curr Mol Med 2015; 15:674-82. [PMID: 26321752 PMCID: PMC5405808 DOI: 10.2174/1566524015666150831141307] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 07/20/2015] [Accepted: 08/24/2015] [Indexed: 12/23/2022]
Abstract
Xerostomia, or chronic dry mouth, is a common syndrome caused by a lack of saliva that can lead to severe eating difficulties, dental caries and oral candida infections. The prevalence of xerostomia increases with age and affects approximately 30% of people aged 65 or older. Given the large numbers of sufferers, and the potential increase in incidence given our aging population, it is important to understand the complex mechanisms that drive hyposalivation and the consequences for the dentition and oral mucosa. From this study we propose the Fgf10 +/- mouse as a model to investigate xerostomia. By following embryonic salivary gland development, in vivo and in vitro, we show that a reduction in Fgf10 causes a delay in branching of salivary glands. This leads to hypoplasia of the glands, a phenotype that is not rescued postnatally or by adulthood in both male and female Fgf10 +/- mice. Histological analysis of the glands showed no obvious defect in cellular differentiation or acini/ductal arrangements, however there was a significant reduction in their size and weight. Analysis of saliva secretion showed that hypoplasia of the glands led to a significant reduction in saliva production in Fgf10 +/- adults, giving rise to a reduced saliva pellicle in the oral cavity of these mice. Mature mice were shown to drink more and in many cases had severe tooth wear. The Fgf10 +/- mouse is therefore a useful model to explore the causes and effects of xerostomia.
Collapse
Affiliation(s)
| | | | | | - A S Tucker
- Department of Craniofacial Development and Stem Cell Biology, Dental Institute, King´s College London, Floor 27 Guy´s Tower, London Bridge, SE1 9RT London, UK.
| |
Collapse
|
109
|
Finley JK, Farmer D, Emmerson E, Cruz Pacheco N, Knox SM. Manipulating the murine lacrimal gland. J Vis Exp 2014:e51970. [PMID: 25490187 DOI: 10.3791/51970] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The lacrimal gland (LG) secretes aqueous tears necessary for maintaining the structure and function of the cornea, a transparent tissue essential for vision. In the human a single LG resides in the orbit above the lateral end of each eye delivering tears to the ocular surface through 3 - 5 ducts. The mouse has three pairs of major ocular glands, the most studied of which is the exorbital lacrimal gland (LG) located anterior and ventral to the ear. Similar to other glandular organs, the LG develops through the process of epithelial branching morphogenesis in which a single epithelial bud within a condensed mesenchyme undergoes multiple rounds of bud and duct formation to form an intricate interconnected network of secretory acini and ducts. This elaborate process has been well documented in many other epithelial organs such as the pancreas and salivary gland. However, the LG has been much less explored and the mechanisms controlling morphogenesis are poorly understood. We suspect that this under-representation as a model system is a consequence of the difficulties associated with finding, dissecting and culturing the LG. Thus, here we describe dissection techniques for harvesting embryonic and post-natal LG and methods for ex vivo culture of the tissue.
Collapse
Affiliation(s)
- Jennifer K Finley
- Craniofacial and Mesenchymal Biology, Cell and Tissue Biology, University of California San Francisco
| | - D'Juan Farmer
- Craniofacial and Mesenchymal Biology, Cell and Tissue Biology, University of California San Francisco
| | - Elaine Emmerson
- Craniofacial and Mesenchymal Biology, Cell and Tissue Biology, University of California San Francisco
| | - Noel Cruz Pacheco
- Craniofacial and Mesenchymal Biology, Cell and Tissue Biology, University of California San Francisco
| | - Sarah M Knox
- Craniofacial and Mesenchymal Biology, Cell and Tissue Biology, University of California San Francisco;
| |
Collapse
|
110
|
Nedvetsky PI, Emmerson E, Finley JK, Ettinger A, Cruz-Pacheco N, Prochazka J, Haddox CL, Northrup E, Hodges C, Mostov KE, Hoffman MP, Knox SM. Parasympathetic innervation regulates tubulogenesis in the developing salivary gland. Dev Cell 2014; 30:449-62. [PMID: 25158854 DOI: 10.1016/j.devcel.2014.06.012] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 04/25/2014] [Accepted: 06/17/2014] [Indexed: 12/11/2022]
Abstract
A fundamental question in development is how cells assemble to form a tubular network during organ formation. In glandular organs, tubulogenesis is a multistep process requiring coordinated proliferation, polarization and reorganization of epithelial cells to form a lumen, and lumen expansion. Although it is clear that epithelial cells possess an intrinsic ability to organize into polarized structures, the mechanisms coordinating morphogenetic processes during tubulogenesis are poorly understood. Here, we demonstrate that parasympathetic nerves regulate tubulogenesis in the developing salivary gland. We show that vasoactive intestinal peptide (VIP) secreted by the innervating ganglia promotes ductal growth, leads to the formation of a contiguous lumen, and facilitates lumen expansion through a cyclic AMP/protein kinase A (cAMP/PKA)-dependent pathway. Furthermore, we provide evidence that lumen expansion is independent of apoptosis and involves the cystic fibrosis transmembrane conductance regulator (CFTR), a cAMP-regulated Cl(-) channel. Thus, parasympathetic innervation coordinates multiple steps in tubulogenesis during organogenesis.
Collapse
Affiliation(s)
- Pavel I Nedvetsky
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Elaine Emmerson
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jennifer K Finley
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Andreas Ettinger
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Noel Cruz-Pacheco
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jan Prochazka
- Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Candace L Haddox
- National Institute of Dental and Craniofacial Research, Bethesda, MD 20892, USA
| | - Emily Northrup
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Craig Hodges
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Keith E Mostov
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Matthew P Hoffman
- National Institute of Dental and Craniofacial Research, Bethesda, MD 20892, USA
| | - Sarah M Knox
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
111
|
Wong HLX, Cao R, Jin G, Ming Chan K, Cao Y, zhou Z. When MT1-MMP meets ADAMs. Cell Cycle 2014; 11:2793-8. [DOI: 10.4161/cc.20949] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
112
|
Shamir ER, Ewald AJ. Three-dimensional organotypic culture: experimental models of mammalian biology and disease. Nat Rev Mol Cell Biol 2014; 15:647-64. [PMID: 25237826 PMCID: PMC4352326 DOI: 10.1038/nrm3873] [Citation(s) in RCA: 532] [Impact Index Per Article: 48.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Mammalian organs are challenging to study as they are fairly inaccessible to experimental manipulation and optical observation. Recent advances in three-dimensional (3D) culture techniques, coupled with the ability to independently manipulate genetic and microenvironmental factors, have enabled the real-time study of mammalian tissues. These systems have been used to visualize the cellular basis of epithelial morphogenesis, to test the roles of specific genes in regulating cell behaviours within epithelial tissues and to elucidate the contribution of microenvironmental factors to normal and disease processes. Collectively, these novel models can be used to answer fundamental biological questions and generate replacement human tissues, and they enable testing of novel therapeutic approaches, often using patient-derived cells.
Collapse
Affiliation(s)
- Eliah R Shamir
- Departments of Cell Biology and Oncology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, 855 North Wolfe Street, Baltimore, Maryland 21205, USA
| | - Andrew J Ewald
- Departments of Cell Biology and Oncology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, 855 North Wolfe Street, Baltimore, Maryland 21205, USA
| |
Collapse
|
113
|
Satoh K, Senpuku H, Sugiya H. Involvement of E2f1 deficiency in salivary gland hypofunction: A review of studies of E2f1-deficient NOD/SCID mice. J Oral Biosci 2014. [DOI: 10.1016/j.job.2014.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
114
|
Hino R, Futagi M, Yamada A, Arakaki M, Saito K, Sugawara Y, Ono M, Fukumoto E, Nakamura T, Fukumoto S. Establishment of ex vivo mucocele model using salivary gland organ culture. PEDIATRIC DENTAL JOURNAL 2014. [DOI: 10.1016/j.pdj.2014.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
115
|
Peters SB, Naim N, Nelson DA, Mosier AP, Cady NC, Larsen M. Biocompatible tissue scaffold compliance promotes salivary gland morphogenesis and differentiation. Tissue Eng Part A 2014; 20:1632-42. [PMID: 24410370 PMCID: PMC4029047 DOI: 10.1089/ten.tea.2013.0515] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 12/12/2013] [Indexed: 12/14/2022] Open
Abstract
Substrate compliance is reported to alter cell phenotype, but little is known about the effects of compliance on cell development within the context of a complex tissue. In this study, we used 0.48 and 19.66 kPa polyacrylamide gels to test the effects of the substrate modulus on submandibular salivary gland development in culture and found a significant decrease in branching morphogenesis in explants grown on the stiff 19.66 kPa gels relative to those grown on the more physiologically compliant 0.48 kPa gels. While proliferation and apoptosis were not affected by the substrate modulus, tissue architecture and epithelial acinar cell differentiation were profoundly perturbed by aberrant, high stiffness. The glands cultured on 0.48 kPa gels were similar to developing glands in morphology and expression of the differentiation markers smooth muscle alpha-actin (SM α-actin) in developing myoepithelial cells and aquaporin 5 (AQP5) in proacinar cells. At 19.66 kPa, however, tissue morphology and the expression and distribution of SM α-actin and AQP5 were disrupted. Significantly, aberrant gland development at 19.66 kPa could be rescued by both mechanical and chemical stimuli. Transfer of glands from 19.66 to 0.48 kPa gels resulted in substantial recovery of acinar structure and differentiation, and addition of exogenous transforming growth factor beta 1 at 19.66 kPa resulted in a partial rescue of morphology and differentiation within the proacinar buds. These results indicate that environmental compliance is critical for organogenesis, and suggest that both mechanical and chemical stimuli can be exploited to promote organ development in the contexts of tissue engineering and organ regeneration.
Collapse
Affiliation(s)
- Sarah B. Peters
- Department of Biological Sciences, University at Albany, State University of New York, Albany, New York
| | - Nyla Naim
- Department of Biological Sciences, University at Albany, State University of New York, Albany, New York
| | - Deirdre A. Nelson
- Department of Biological Sciences, University at Albany, State University of New York, Albany, New York
| | - Aaron P. Mosier
- College of Nanoscale Science and Engineering, University at Albany, State University of New York, Albany, New York
| | - Nathaniel C. Cady
- College of Nanoscale Science and Engineering, University at Albany, State University of New York, Albany, New York
| | - Melinda Larsen
- Department of Biological Sciences, University at Albany, State University of New York, Albany, New York
| |
Collapse
|
116
|
Matsumoto S, Fujii S, Sato A, Ibuka S, Kagawa Y, Ishii M, Kikuchi A. A combination of Wnt and growth factor signaling induces Arl4c expression to form epithelial tubular structures. EMBO J 2014; 33:702-718. [PMID: 24562386 PMCID: PMC4000088 DOI: 10.1002/embj.201386942] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 01/13/2014] [Accepted: 01/14/2014] [Indexed: 12/17/2022] Open
Abstract
Growth factor-dependent epithelial morphological changes and proliferation are essential for the formation of tubular structures, but the underlying molecular mechanisms are poorly understood. Co-stimulation with Wnt3a and epidermal growth factor (Wnt3a/EGF) induced development of tubes consisting of intestinal epithelial cells by inducing expression of Arl4c, an Arf-like small GTP-binding protein, in three-dimensional culture, while stimulation with Wnt3a or EGF alone did not. Arl4c expression resulted in rearrangement of the cytoskeleton through activation of Rac and inactivation of Rho properly, which promoted cell growth by inducing nuclear translocation of Yes-associated protein and transcriptional co-activator with PDZ-binding motif (YAP/TAZ) in leading cells. Arl4c was expressed in ureteric bud tips and pretubular structures in the embryonic kidney. In an organoid culture assay, Wnt and fibroblast growth factor signaling simultaneously induced elongation and budding of kidney ureteric buds through Arl4c expression. YAP/TAZ was observed in the nucleus of extending ureteric bud tips. Thus, Arl4c expression induced by a combination of growth factor signaling mechanisms is involved in tube formation.
Collapse
Affiliation(s)
- Shinji Matsumoto
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka UniversityOsaka, Japan
| | - Shinsuke Fujii
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka UniversityOsaka, Japan
- Interdisciplinary Program for Biomedical Sciences (IPBS), Institute for Academic Initiatives, Osaka University, Graduate School of MedicineOsaka, Japan
| | - Akira Sato
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka UniversityOsaka, Japan
| | - Souji Ibuka
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka UniversityOsaka, Japan
- Department of Pediatric Surgery, Graduate School of Medicine, Osaka UniversityOsaka, Japan
| | - Yoshinori Kagawa
- Department of Gastrointestinal Surgery, Graduate School of Medicine, Osaka UniversityOsaka, Japan
- Department of Immunology and Cell Biology, Graduate School of Medicine, Osaka UniversityOsaka, Japan
- Japan Science and Technology Agency (JST), CRESTTokyo, Japan
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine, Osaka UniversityOsaka, Japan
- Japan Science and Technology Agency (JST), CRESTTokyo, Japan
| | - Akira Kikuchi
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka UniversityOsaka, Japan
| |
Collapse
|
117
|
Küchler EC, Feng P, Deeley K, Fitzgerald CA, Meyer C, Gorbunov A, Bezamat M, Reis MF, Noel J, Kouzbari MZ, Granjeiro JM, Antunes LS, Antunes LA, de Abreu FV, Costa MC, Tannure PN, Seymen F, Koruyucu M, Patir A, Vieira AR. Fine mapping of locus Xq25.1-27-2 for a low caries experience phenotype. Arch Oral Biol 2014; 59:479-86. [PMID: 24632093 DOI: 10.1016/j.archoralbio.2014.02.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 01/17/2014] [Accepted: 02/15/2014] [Indexed: 10/25/2022]
Abstract
OBJECTIVE The purpose of this study was to fine map the locus Xq25.1-27-2 in order to identify genetic contributors involved in low caries experience. DESIGN Seventy-two families from the Philippines were studied. Caries experience was recorded and genomic DNA extracted from peripheral blood was obtained from all subjects. One hundred and twenty-eight polymorphisms in the locus Xq25.1-27-2, a region that contains 24 genes, were genotyped. Association between caries experience and alleles was tested using the transmission disequilibrium test (TDT). This initial analysis was followed by experiments with DNA samples from 1481 subjects from Pittsburgh, 918 children from Brazil, and 275 children from Turkey in order to follow up the results found in the Filipino families. Chi-square or Fisher's exact tests were used. Sequencing of the coding regions and exon-intron boundaries of MST4 and FGF13 were also performed on 91 women from Pittsburgh. RESULTS Statistically significant association with low caries experience was found for 11 markers in Xq25.1-27-2 in the Filipino families. One marker was in MST4, another marker was in FGF13, and the remaining markers were in intergenic regions. Haplotype analysis also confirmed these results, but the follow up studies with DNA samples from Pittsburgh, Brazil, and Turkey showed associations for a subset of the 11 markers. No coding mutations were identified by sequencing. CONCLUSIONS Our study failed to conclusively demonstrate that genetic factors in Xq25.1-27-2 contribute to caries experience in multiple populations.
Collapse
Affiliation(s)
- Erika C Küchler
- Department of Oral Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Ping Feng
- Guiyang Stomatological Hospital, Guiyang, Guizhou, China.
| | - Kathleen Deeley
- Department of Oral Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Carly A Fitzgerald
- Department of Oral Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Chelsea Meyer
- Department of Oral Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Anastasia Gorbunov
- Department of Oral Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Mariana Bezamat
- Department of Oral Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Maria Fernanda Reis
- Department of Oral Biology, University of Pittsburgh, Pittsburgh, PA, USA; Clinical Research Unit, Fluminense Federal University, Niterói, RJ, Brazil.
| | - Jacqueline Noel
- Department of Oral Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - M Zahir Kouzbari
- Department of Oral Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - José M Granjeiro
- Clinical Research Unit, Fluminense Federal University, Niterói, RJ, Brazil; Directory of Programs, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ, Brazil.
| | - Leonardo S Antunes
- Clinical Research Unit, Fluminense Federal University, Niterói, RJ, Brazil; Pediatric Clinics, School of Dentistry, Fluminense Federal University, Nova Friburgo, RJ, Brazil.
| | - Livia A Antunes
- Pediatric Clinics, School of Dentistry, Fluminense Federal University, Nova Friburgo, RJ, Brazil.
| | - Fernanda Volpe de Abreu
- Pediatric Clinics, School of Dentistry, Fluminense Federal University, Nova Friburgo, RJ, Brazil.
| | - Marcelo C Costa
- Department of Pediatric Dentistry and Orthodontics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Patricia N Tannure
- Veiga de Almeida University, Rio de Janeiro, RJ, Brazil; Discipline of Cariology, School of Dentistry, Salgado de Oliveira University, Niterói, RJ, Brazil.
| | - Figen Seymen
- Department of Pedodontics, Istanbul University, Istanbul, Turkey.
| | - Mine Koruyucu
- Department of Pedodontics, Istanbul University, Istanbul, Turkey.
| | - Asli Patir
- Department of Pedodontics, Medipol Istanbul University, Istanbul, Turkey.
| | - Alexandre R Vieira
- Department of Oral Biology, University of Pittsburgh, Pittsburgh, PA, USA; Center for Craniofacial and Dental Genetics, and Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pediatric Dentistry, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
118
|
Wang X, Qi S, Wang J, Xia D, Qin L, Zheng Z, Wang L, Zhang C, Jin L, Ding G, Wang S, Fan Z. Spatial and temporal expression of c-Kit in the development of the murine submandibular gland. J Mol Histol 2014; 45:381-9. [PMID: 24554067 DOI: 10.1007/s10735-014-9570-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Accepted: 02/11/2014] [Indexed: 02/06/2023]
Abstract
The c-Kit pathway is important in the development of many mammalian cells and organs and is indispensable for the development of hematopoiesis, melanocytes, and primordial germ cells. Loss-of-function mutations in c-Kit lead to perinatal death in mouse embryos. Previously, c-Kit has been used as one of salivary epithelial stem or progenitor cell markers in mouse, its specific temporo-spatial expression pattern and function in developing murine submandibular gland (SMG) is still unclear. Here we used quantitative real-time PCR, in situ hybridization, and immunohistochemistry analysis to detect c-Kit expression during the development of the murine SMG. We found that c-Kit was expressed in the epithelia of developing SMGs from embryonic day 11.5 (E11.5; initial bud stage) to postnatal day 90 (P90; when the SMG is completely mature). c-Kit expression in the end bud epithelium increased during prenatal development and then gradually decreased after birth until its expression was undetectable in mature acini at P30. Moreover, c-Kit was expressed in the SMG primordial cord at the initial bud, pseudoglandular, canacular, and terminal end bud stages. c-Kit was also expressed in the presumptive ductal cells adjacent to the developing acini. By the late terminal end bud stage on P14, c-Kit expression could not be detected in ductal cells. However, c-Kit expression was detected in ductal cells at P30, and its expression had increased dramatically at P90. Taken together, these findings describe the spatial and temporal expression pattern of c-Kit in the developing murine SMG and suggest that c-Kit may play roles in epithelial histo-morphogenesis and in ductal progenitor cell homeostasis in the SMG.
Collapse
Affiliation(s)
- Xuejiu Wang
- Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, 100050, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
119
|
Filant J, DeMayo FJ, Pru JK, Lydon JP, Spencer TE. Fibroblast growth factor receptor two (FGFR2) regulates uterine epithelial integrity and fertility in mice. Biol Reprod 2014; 90:7. [PMID: 24227756 PMCID: PMC7289345 DOI: 10.1095/biolreprod.113.114496] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 10/29/2013] [Accepted: 11/06/2013] [Indexed: 12/20/2022] Open
Abstract
Fibroblast growth factors (FGFs) and their receptors (FGFRs) regulate luminal epithelial (LE) cell proliferation in the adult mouse uterus. This study tested the hypothesis that FGFR2 has a biological role in postnatal development and function of the uterus by conditionally deleting Fgfr2 after birth using progesterone receptor (Pgr)-Cre mice. Adult Fgfr2 mutant female mice were initially subfertile and became infertile with increasing parity. No defects in uterine gland development were observed in conditional Fgfr2 mutant mice. In the adult, Fgfr2 mutant mice possessed a histologically normal reproductive tract with the exception of the uterus. The LE of the Fgfr2 mutant uterus was stratified, but no obvious histological differences were observed in the glandular epithelium, stroma, or myometrium. Within the stratified LE, cuboidal basal cells were present and positive for basal cell markers (KRT14 and TRP63). Nulliparous bred Fgfr2 mutants contained normal numbers of blastocysts on Day 3.5 postmating, but the number of embryo implantation sites was substantially reduced on Day 5.5 postmating. These results support the idea that loss of FGFR2 in the uterus after birth alters its development, resulting in LE stratification and peri-implantation pregnancy loss.
Collapse
Affiliation(s)
- Justyna Filant
- Center for Reproductive Biology, Department of Animal Sciences, Washington State University, Pullman, Washington
| | | | | | | | | |
Collapse
|
120
|
Kera H, Yuki S, Nogawa H. FGF7 signals are relayed to autocrine EGF family growth factors to induce branching morphogenesis of mouse salivary epithelium. Dev Dyn 2013; 243:552-9. [DOI: 10.1002/dvdy.24097] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 10/25/2013] [Accepted: 11/11/2013] [Indexed: 12/26/2022] Open
Affiliation(s)
- Hayashi Kera
- Department of Biology; Graduate School of Science; Chiba University; Chiba Japan
| | - Satoshi Yuki
- Department of Biology; Graduate School of Science; Chiba University; Chiba Japan
| | - Hiroyuki Nogawa
- Department of Biology; Graduate School of Science; Chiba University; Chiba Japan
| |
Collapse
|
121
|
Patel VN, Hoffman MP. Salivary gland development: a template for regeneration. Semin Cell Dev Biol 2013; 25-26:52-60. [PMID: 24333774 DOI: 10.1016/j.semcdb.2013.12.001] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 11/26/2013] [Accepted: 12/02/2013] [Indexed: 12/16/2022]
Abstract
The mammalian salivary gland develops as a highly branched structure designed to produce and secrete saliva. This review will focus on research on mouse submandibular gland development and the translation of this basic research toward therapy for patients suffering from salivary hypofunction. Here we review the most recent literature that has enabled a better understanding of the mechanisms of salivary gland development. Additionally, we discuss approaches proposed to restore salivary function using gene and cell-based therapy. Increasing our understanding of the developmental mechanisms involved during development is critical to design effective therapies for regeneration and repair of damaged glands.
Collapse
Affiliation(s)
- Vaishali N Patel
- Matrix and Morphogenesis Section, Laboratory of Cell and Developmental Biology, NIDCR, NIH, Bethesda, MD 20892, United States
| | - Matthew P Hoffman
- Matrix and Morphogenesis Section, Laboratory of Cell and Developmental Biology, NIDCR, NIH, Bethesda, MD 20892, United States.
| |
Collapse
|
122
|
Ray S, Yuan D, Dhulekar N, Oztan B, Yener B, Larsen M. Cell-based multi-parametric model of cleft progression during submandibular salivary gland branching morphogenesis. PLoS Comput Biol 2013; 9:e1003319. [PMID: 24277996 PMCID: PMC3836695 DOI: 10.1371/journal.pcbi.1003319] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 09/18/2013] [Indexed: 12/12/2022] Open
Abstract
Cleft formation during submandibular salivary gland branching morphogenesis is the critical step initiating the growth and development of the complex adult organ. Previous experimental studies indicated requirements for several epithelial cellular processes, such as proliferation, migration, cell-cell adhesion, cell-extracellular matrix (matrix) adhesion, and cellular contraction in cleft formation; however, the relative contribution of each of these processes is not fully understood since it is not possible to experimentally manipulate each factor independently. We present here a comprehensive analysis of several cellular parameters regulating cleft progression during branching morphogenesis in the epithelial tissue of an early embryonic salivary gland at a local scale using an on lattice Monte-Carlo simulation model, the Glazier-Graner-Hogeweg model. We utilized measurements from time-lapse images of mouse submandibular gland organ explants to construct a temporally and spatially relevant cell-based 2D model. Our model simulates the effect of cellular proliferation, actomyosin contractility, cell-cell and cell-matrix adhesions on cleft progression, and it was used to test specific hypotheses regarding the function of these parameters in branching morphogenesis. We use innovative features capturing several aspects of cleft morphology and quantitatively analyze clefts formed during functional modification of the cellular parameters. Our simulations predict that a low epithelial mitosis rate and moderate level of actomyosin contractility in the cleft cells promote cleft progression. Raising or lowering levels of contractility and mitosis rate resulted in non-progressive clefts. We also show that lowered cell-cell adhesion in the cleft region and increased cleft cell-matrix adhesions are required for cleft progression. Using a classifier-based analysis, the relative importance of these four contributing cellular factors for effective cleft progression was determined as follows: cleft cell contractility, cleft region cell-cell adhesion strength, epithelial cell mitosis rate, and cell-matrix adhesion strength. Branching morphogenesis is a complex and dynamic embryonic process that creates the structure of many adult organs, including the salivary gland. During this process, many cellular changes occur in the epithelial cells, including changes in cell-cell adhesions, cell-extracellular matrix (matrix) adhesions, cell proliferation, and cellular contraction, resulting in formation of clefts in the epithelial cells of the organ. A comprehensive understanding of the relative contributions of these cellular processes has crucial therapeutic implications for organ regeneration and functional restoration of organ structure in diseased salivary glands. Here, we have developed a cell-based model of cleft progression and simulated cleft progression under conditions of altered cell-cell adhesions, cellular contractility, cell-matrix adhesion and cell proliferation to identify the optimum cellular conditions that cause clefts to progress. The model predicts that cleft progression requires a moderate level of cleft cell contractility, a low epithelial proliferation rate, reduced cell-cell adhesion strength in the cleft and high cell-matrix adhesion strength also in the cleft region. The results of our classification analysis demonstrate that cellular contractility in the cleft cells has a significant effect on cleft progression, followed by cell-cell adhesion strength, rate of cell proliferation, and strength of cell-matrix adhesion energies.
Collapse
Affiliation(s)
- Shayoni Ray
- Department of Biological Sciences, University at Albany, State University of New York, Albany, New York, United States of America
| | - Daniel Yuan
- Department of Computer Science, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Nimit Dhulekar
- Department of Computer Science, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Basak Oztan
- Department of Computer Science, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Bülent Yener
- Department of Computer Science, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Melinda Larsen
- Department of Biological Sciences, University at Albany, State University of New York, Albany, New York, United States of America
- * E-mail:
| |
Collapse
|
123
|
Wells KL, Gaete M, Matalova E, Deutsch D, Rice D, Tucker AS. Dynamic relationship of the epithelium and mesenchyme during salivary gland initiation: the role of Fgf10. Biol Open 2013; 2:981-9. [PMID: 24167707 PMCID: PMC3798193 DOI: 10.1242/bio.20135306] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 07/07/2013] [Indexed: 12/19/2022] Open
Abstract
Salivary glands provide an excellent model for the study of epithelial-mesenchymal interactions. We have looked at the interactions involved in the early initiation and development of murine salivary glands using classic recombination experiments and knockout mice. We show that salivary gland epithelium, at thickening and initial bud stages, is able to direct salivary gland development in non-gland pharyngeal arch mesenchyme at early stages. The early salivary gland epithelium is therefore able to induce gland development in non-gland tissue. This ability later shifts to the mesenchyme, with non-gland epithelium, such as from the limb bud, able to form a branching gland when combined with pseudoglandular stage gland mesenchyme. This shift appears to involve Fgf signalling, with signals from the epithelium inducing Fgf10 in the mesenchyme. Fgf10 then signals back to the epithelium to direct gland down-growth and bud development. These experiments highlight the importance of epithelial-mesenchymal signalling in gland initiation, controlling where, when and how many salivary glands form.
Collapse
Affiliation(s)
- Kirsty L. Wells
- Department of Craniofacial Development and Stem Cell Biology, King's College London, Floor 27, Guy's Tower, London Bridge, London SE1 9RT, UK
| | - Marcia Gaete
- Department of Craniofacial Development and Stem Cell Biology, King's College London, Floor 27, Guy's Tower, London Bridge, London SE1 9RT, UK
- Faculty of Medicine, Pontificia Universidad Católica de Chile, Alameda 340, Santiago, 8331150, Chile
| | - Eva Matalova
- Institute of Animal Physiology and Genetics, v.v.i., Academy of Sciences of the Czech Republic, 602 00, Brno, Czech Republic
- Department of Physiology, University of Veterinary and Pharmaceutical Sciences, 612 42 Brno, Czech Republic
| | - Danny Deutsch
- Dental Research Laboratory, Institute of Dental Sciences, Hebrew University, Hadassah, Faculty of Dental Medicine, Jerusalem 91120, Israel
| | - David Rice
- Department of Orthodontics, Institute of Dentistry, University of Helsinki, Helsinki 00014, Finland
- Oral and Maxillofacial Diseases, Helsinki University Central Hospital, Helsinki 00290, Finland
| | - Abigail S. Tucker
- Department of Craniofacial Development and Stem Cell Biology, King's College London, Floor 27, Guy's Tower, London Bridge, London SE1 9RT, UK
| |
Collapse
|
124
|
Xiao N, Cao H, Chen CH, Kong CS, Ali R, Chan C, Sirjani D, Graves E, Koong A, Giaccia A, Mochly-Rosen D, Le QT. A novel aldehyde dehydrogenase-3 activator (Alda-89) protects submandibular gland function from irradiation without accelerating tumor growth. Clin Cancer Res 2013; 19:4455-64. [PMID: 23812668 DOI: 10.1158/1078-0432.ccr-13-0127] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
PURPOSE To determine the effect of Alda-89 (an ALDH3 activitor) on (i) the function of irradiated (radiotherapy) submandibular gland (SMG) in mice, (ii) its toxicity profile, and (iii) its effect on the growth of head and neck cancer (HNC) in vitro and in vivo. EXPERIMENTAL DESIGN Adult mice were infused with Alda-89 or vehicle before, during, and after radiotherapy. Saliva secretion was monitored weekly. Hematology, metabolic profile, and postmortem evaluation for toxicity were examined at the time of sacrifice. Alda-89 or vehicle was applied to HNC cell lines in vitro, and severe combined immunodeficient (SCID) mice transplanted with HNC in vivo with or without radiation; HNC growth was monitored. The ALDH3A1 and ALDH3A2 protein expression was evaluated in 89 patients with HNC and correlated to freedom from relapse (FFR) and overall survival (OS). RESULTS Alda-89 infusion significantly resulted in more whole saliva production and a higher percentage of preserved acini after radiotherapy compared with vehicle control. There was no difference in the complete blood count, metabolic profile, and major organ morphology between the Alda-89 and vehicle groups. Compared with vehicle control, Alda-89 treatment neither accelerated HNC cell proliferation in vitro, nor did it affect tumor growth in vivo with or without radiotherapy. Higher expression of ALDH3A1 or ALDH3A2 was not significantly associated with worse FFR or OS in either human papillomavirus (HPV)-positive or HPV-negative group. CONCLUSION Alda-89 preserves salivary function after radiotherapy without affecting HNC growth or causing measurable toxicity in mice. It is a promising candidate to mitigate radiotherapy-related xerostomia.
Collapse
Affiliation(s)
- Nan Xiao
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
125
|
Parasympathetic stimulation improves epithelial organ regeneration. Nat Commun 2013; 4:1494. [PMID: 23422662 PMCID: PMC3582394 DOI: 10.1038/ncomms2493] [Citation(s) in RCA: 146] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 01/15/2013] [Indexed: 12/25/2022] Open
Abstract
Parasympathetic nerves are a vital component of the progenitor cell niche during development, maintaining a pool of progenitors for organogenesis. Injured adult organs do not regenerate after parasympathectomy, and there are few treatments to improve organ regeneration, particularly after damage by therapeutic irradiation. Here we show that restoring parasympathetic function with the neurotrophic factor neurturin increases epithelial organ regeneration after damage. We use mouse salivary gland explant culture containing fluorescently-labeled progenitors, and injure the tissue with irradiation. The progenitors survive, parasympathetic function is diminished, and epithelial apoptosis reduces expression of neurturin, which increases neuronal apoptosis. Treatment with neurturin reduces neuronal apoptosis, restores parasympathetic function, and increases epithelial regeneration. Furthermore adult human salivary glands damaged by irradiation also have reduced parasympathetic innervation. We propose that neurturin will protect the parasympathetic nerves from damage and improve organ regeneration. This concept may be applicable for other organs where parasympathetic innervation influences their function.
Collapse
|
126
|
Expression and functions of fibroblast growth factor 10 in the mouse mammary gland. Int J Mol Sci 2013; 14:4094-105. [PMID: 23434672 PMCID: PMC3588087 DOI: 10.3390/ijms14024094] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 01/15/2013] [Accepted: 02/05/2013] [Indexed: 01/14/2023] Open
Abstract
Fibroblast growth factor 10 (FGF10) is important as a mesenchymal mediator of epithelial growth and morphogenesis. In this study, the expression and localization of the FGF10 protein were detected by laser scanning confocal microscopy during mouse postnatal mammary gland development. Mammary explants were cultured to investigate the functions of FGF10. The results revealed that FGF10 localizes mainly in the mesenchyme near the ductal epithelial cells and the alveolar epithelial cells of the mammary gland. Peak FGF10 expression levels were observed at lactation day 10. FGF10 induced FGFR2-IIIb expression in the mammary epithelium, except in virgin or pregnant mice. FGF10 promoted the proliferation of mammary gland epithelial cells and reduced apoptosis. FGF10 is important during the mouse mammary gland growth, development, and reconstruction, and its effects are mediated by FGFR2-IIIb.
Collapse
|
127
|
He D, Ujjal K. Bhawal, Hamada N, Kuboyama N, Abiko Y, Arakawa H. Low Level Fluoride Stimulates Epithelial-Mesenchymal Interaction in Oral Mucosa. J HARD TISSUE BIOL 2013. [DOI: 10.2485/jhtb.22.59] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
128
|
Yang TL, Lin L, Hsiao YC, Lee HW, Young TH. Chitosan Biomaterials Induce Branching Morphogenesis in a Model of Tissue-Engineered Glandular Organs in Serum-Free Conditions. Tissue Eng Part A 2012; 18:2220-30. [DOI: 10.1089/ten.tea.2011.0527] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Tsung-Lin Yang
- Department of Otolaryngology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| | - Lin Lin
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Ya-Chuan Hsiao
- Department of Ophthalmology, Zhongxing Branch, Taipei City Hospital, Taipei, Taiwan
- Department of Ophthalmology, College of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Hao-Wei Lee
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Tai-Horng Young
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
129
|
Koyama N, Hayashi T, Mizukoshi K, Matsumoto T, Gresik EW, Kashimata M. Extracellular regulated kinase5 is expressed in fetal mouse submandibular glands and is phosphorylated in response to epidermal growth factor and other ligands of the ErbB family of receptors. Dev Growth Differ 2012; 54:801-8. [PMID: 23078124 DOI: 10.1111/dgd.12008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 09/09/2012] [Accepted: 09/10/2012] [Indexed: 12/28/2022]
Abstract
Growth factors and their receptors regulate development of many organs through activation of multiple intracellular signaling cascades including a mitogen-activated protein kinase (MAPK). Extracellular regulated kinases (ERK)1/2, classic MAPK family members, are expressed in fetal mouse submandibular glands (SMG), and stimulate branching morphogenesis. ERK5, also called big mitogen-activated protein kinase 1, was recently found as a new member of MAPK super family, and its biological roles are still largely unknown. In this study, we investigated the expression and function of ERK5 in developing fetal mouse SMGs. Western blotting analysis showed that the expression pattern of ERK5 was different from the pattern of ERK1/2 in developing fetal SMGs. Both ERK1/2 and ERK5 were phosphorylated after exposure to ligands of the ErbB family of receptor tyrosine kinases (RTKs). Phosphorylation of ERK1/2 was strongly induced by epidermal growth factor (EGF) in SMG rudiments at embryonic day 14 (E14), E16 and E18. However, ERK5 phosphorylation induced by EGF was clearly observed at E14 and E16, but not at E18. Branching morphogenesis of cultured E13 SMG rudiments was strongly suppressed by administration of U0126, an inhibitor for ERK1/2 activation, whereas the phosphorylation of ERK5 was not inhibited by U0126. BIX02188, a specific inhibitor for ERK5 activation, also inhibited branching morphogenesis in cultured SMG rudiments. These results show that EGF-responsive ERK5 is expressed in developing fetal mouse SMG, and suggest that both ERK1/2 and ERK5 signaling cascades might play an important role in the regulation of branching morphogenesis.
Collapse
Affiliation(s)
- Noriko Koyama
- Department of Pharmacology, Asahi University School of Dentistry, Hozumi, Mizuho, Gifu, 501-0296, Japan
| | | | | | | | | | | |
Collapse
|
130
|
Alam DA, Sala FG, Baptista S, Galzote R, Danopoulos S, Tiozzo C, Gage P, Grikscheit T, Warburton D, Frey MR, Bellusci S. FGF9-Pitx2-FGF10 signaling controls cecal formation in mice. Dev Biol 2012; 369:340-8. [PMID: 22819677 PMCID: PMC3725282 DOI: 10.1016/j.ydbio.2012.07.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 06/19/2012] [Accepted: 07/10/2012] [Indexed: 01/09/2023]
Abstract
Fibroblast growth factor (FGF) signaling to the epithelium and mesenchyme mediated by FGF10 and FGF9, respectively, controls cecal formation during embryonic development. In particular, mesenchymal FGF10 signals to the epithelium via FGFR2b to induce epithelial cecal progenitor cell proliferation. Yet the precise upstream mechanisms controlling mesenchymal FGF10 signaling are unknown. Complete deletion of Fgf9 as well as of Pitx2, a gene encoding a homeobox transcription factor, both lead to cecal agenesis. Herein, we used mouse genetic approaches to determine the precise contribution of the epithelium and/or mesenchyme tissue compartments in this process. Using tissue compartment specific Fgf9 versus Pitx2 loss of function approaches in the gut epithelium and/or mesenchyme, we determined that FGF9 signals to the mesenchyme via Pitx2 to induce mesenchymal Fgf10 expression, which in turn leads to epithelial cecal bud formation.
Collapse
MESH Headings
- Animals
- Base Sequence
- Cecum/abnormalities
- Cecum/embryology
- Cecum/metabolism
- Cell Proliferation
- DNA Primers/genetics
- Epithelial Cells/cytology
- Epithelial Cells/metabolism
- Female
- Fibroblast Growth Factor 10/deficiency
- Fibroblast Growth Factor 10/genetics
- Fibroblast Growth Factor 10/metabolism
- Fibroblast Growth Factor 9/deficiency
- Fibroblast Growth Factor 9/genetics
- Fibroblast Growth Factor 9/metabolism
- Gene Expression Regulation, Developmental
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Male
- Mesoderm/embryology
- Mesoderm/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Mutant Strains
- Mice, Transgenic
- Models, Biological
- Pregnancy
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 2/metabolism
- Signal Transduction
- Transcription Factors/deficiency
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Homeobox Protein PITX2
Collapse
Affiliation(s)
- Denise Al Alam
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Frederic G Sala
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Sheryl Baptista
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Rosanna Galzote
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Soula Danopoulos
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Caterina Tiozzo
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Philip Gage
- University of Michigan Kellogg Eye Center, 1000 Wall Street, Ann Arbor, MI, USA
| | - Tracy Grikscheit
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - David Warburton
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Mark R Frey
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Saverio Bellusci
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- Excellence Cluster in Cardio-Pulmonary Systems, University of Giessen Lung Center, Department of Internal Medicine II, Klinikstrasse 36, 35392 Giessen, Germany
| |
Collapse
|
131
|
Abstract
PURPOSE This study aimed to review and discuss the utility of the Fgfr2 (W290R) mouse mutant as a model of human Crouzon syndrome. METHODS A review of current and past scientific literature on Fibroblast Growth Factor Receptor-2 (FGFR2) protein domain structure, FGFR mutations associated with human Crouzon syndrome, and phenotypic and molecular changes combined with recent observations and experimental data of the Fgfr2 (W290R) mouse mutant was conducted. A comparison of the Fgfr2 (W290R) mouse mutant with another mouse model of Crouzon syndrome, Fgfr2 (C342R) mouse mutant, was also performed. Finally, possible future research directions using the Fgfr2 (W290R) mutant mice were discussed. RESULTS The Fgfr2 (W290R) heterozygous mouse exhibits defects characteristic of human Crouzon syndrome. At the molecular level, the defects observed in the mouse mutant are due to the dysregulation of signaling of both the IIIb and IIIc isoforms of Fgfr2. The involvement of the IIIb isoform of FGFR2 in the etiopathology of Crouzon syndrome is a novel finding in the craniosynostosis literature field. Dysregulated signaling of both IIIb and IIIc isoforms causes a broad spectrum of changes that explain some of the defects observed clinically in humans. Several of the defects observed in the Fgfr2 (W290R) homozygous mouse mutant are attributable to a loss-of-function mechanism in contrast to the frequently reported gain-of-function receptor function associated with mutated FGF receptors in craniosynostosis. CONCLUSIONS The Fgfr2 ( W290R ) mouse model can be used as a model system to further investigate the cellular, molecular, and biochemical mechanisms of Crouzon syndrome.
Collapse
|
132
|
Oron G, Fisch B, Zhang XY, Gabbay-Benziv R, Kessler-Icekson G, Krissi H, Ben-Haroush A, Ao A, Abir R. Fibroblast growth factor 10 in human ovaries. Reprod Biomed Online 2012; 25:396-401. [PMID: 22877940 DOI: 10.1016/j.rbmo.2012.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2012] [Revised: 06/12/2012] [Accepted: 07/11/2012] [Indexed: 11/28/2022]
Abstract
The expression of fibroblast growth factor 10 (FGF-10) has not been studied in human ovarian cortical follicles. The aim of the present study was to investigate the expression of FGF-10 in preantral follicles from fetuses, girls and women. Ovarian samples were obtained from 14 human fetuses at 21-33 gestational weeks and from 35 girls and women aged 5-39 years. The specimens were prepared for detection of the FGF-10 protein by immunohistochemistry. Reverse-transcription PCR was applied to ovarian extracts to identify FGF-10 mRNA transcripts. In fetal tissue, the FGF-10 protein was detected in oocytes in 50% of the samples and in granulosa cells in 30%. In ovarian tissue from girls and women, the FGF-10 protein was detected in oocytes and granulosa cells in all samples. FGF-10 mRNA transcripts were present in all adult and fetal samples tested. The identification of FGF-10 at both the protein and mRNA levels suggests that FGF-10 may contribute to human preantral follicle development.
Collapse
Affiliation(s)
- Galia Oron
- Infertility and IVF Unit, Hospital for Women, Rabin Medical Center, Petach Tikva 49100, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
133
|
Gasperin BG, Ferreira R, Rovani MT, Santos JT, Buratini J, Price CA, Gonçalves PBD. FGF10 inhibits dominant follicle growth and estradiol secretion in vivo in cattle. Reproduction 2012; 143:815-23. [DOI: 10.1530/rep-11-0483] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Fibroblast growth factors (FGFs) are involved in paracrine control of follicle development. It was previously demonstrated that FGF10 decreases estradiol (E2) secretion in granulosa cell culture and that theca cell FGF10 mRNA expression is decreased in healthy follicles from abattoir ovaries. The main objectives of this study were to evaluate FGF10 and FGFR2b mRNA expression during follicular development in vivo, to evaluate the effect of FGF10 on follicle growth using Bos taurus taurus cows as a model, and to gain more insight into the mechanisms through which FGF10 inhibits steroidogenesis. Messenger RNA encoding both FGF10 and FGFR2b (main FGF10 receptor) was significantly more expressed in subordinate follicles (SFs) than in dominant follicles (DFs). The intrafollicular injection of FGF10 into the largest growing follicle at 7–8 mm in diameter interrupted the DF growth in a dose-dependent manner (11±0.4, 8.3±1 and 5.9±0.3 mm for 0, 0.1, and 1 μg/ml FGF10, respectively, at 72 h after treatment; P<0.05). In a third experiment, follicles were obtained 24 h after FGF10 (1 μg/ml) or PBS treatment through ovariectomy. In theca cells, FGF10 treatment did not affect mRNA encoding steroidogenic enzymes, LHCGR and IGFBPs, but significantly upregulated FGF10 mRNA expression. The expression of CYP19A1 mRNA in granulosa cells was downregulated by FGF10 treatment, which was accompanied by a 50-fold decrease in E2 production, and decreased cyclin D2 mRNA. These results have shown that FGF10 and its receptor FGFR2b are more expressed in SFs and provide solid in vivo evidence that FGF10 acts as an important regulator of follicular growth in cattle.
Collapse
|
134
|
Tian E, Hoffman MP, Ten Hagen KG. O-glycosylation modulates integrin and FGF signalling by influencing the secretion of basement membrane components. Nat Commun 2012; 3:869. [PMID: 22643896 PMCID: PMC3533247 DOI: 10.1038/ncomms1874] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 04/27/2012] [Indexed: 01/08/2023] Open
Abstract
Extracellular microenvironments have crucial roles in modulating cell interactions during development. Here we discover that a conserved protein modification (O-glycosylation) influences extracellular matrix composition during mammalian organogenesis, affecting integrin signalling and fibroblast growth factor-mediated cell proliferation. Specifically, mice deficient for an enzyme (Galnt1) that adds sugars to proteins during early stages of organogenesis resulted in intracellular accumulation of major basement membrane proteins and endoplasmic reticulum stress, with resultant effects on fibroblast growth factor signalling, epithelial cell proliferation and organ growth. Exogenous addition of basement membrane components rescued fibroblast growth factor signalling and the growth defects in a β1-integrin-dependent manner. Our work demonstrates for the first time that O-glycosylation influences the composition of the extracellular matrix during mammalian organ development, influencing specific aspects of the endoplasmic reticulum stress response, cell signalling, cell proliferation and organ growth. Our work provides insight into the role of this conserved protein modification in both development and disease.
Collapse
Affiliation(s)
- E Tian
- Developmental Glycobiology Unit, Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892-4370, USA
| | - Matthew P. Hoffman
- Matrix and Morphogenesis Section, Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892-4370, USA
| | - Kelly G. Ten Hagen
- Developmental Glycobiology Unit, Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892-4370, USA
| |
Collapse
|
135
|
Kumar M, Chapman SC. Cloning and expression analysis of Fgf5, 6 and 7 during early chick development. Gene Expr Patterns 2012; 12:245-53. [PMID: 22634565 DOI: 10.1016/j.gep.2012.05.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Revised: 05/16/2012] [Accepted: 05/17/2012] [Indexed: 02/02/2023]
Abstract
FGFs with similar sequences can play different roles depending on the model organisms examined. Determining these roles requires knowledge of spatio-temporal Fgf gene expression patterns. In this study, we report the cloning of chick Fgf5, 6 and 7, and examine their gene expression patterns by whole mount in situ hybridization. We show that Fgf5's spatio-temporally restricted expression pattern indicates a potentially novel role during inner ear development. Fgf6 and Fgf7, although belonging to different subfamilies with diverged sequences, are expressed in similar patterns within the mesoderm. Alignment of protein sequences and phylogenetic analysis demonstrate that FGF5 and FGF6 are highly conserved between chick, human, mouse and zebrafish. FGF7 is similarly conserved except for the zebrafish, which has considerably diverged.
Collapse
Affiliation(s)
- Megha Kumar
- Clemson University, Biological Sciences, Long Hall, Clemson, SC 29634, USA
| | | |
Collapse
|
136
|
Zhu W, Nelson CM. PI3K signaling in the regulation of branching morphogenesis. Biosystems 2012; 109:403-11. [PMID: 22525052 DOI: 10.1016/j.biosystems.2012.04.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2012] [Revised: 04/03/2012] [Accepted: 04/11/2012] [Indexed: 11/25/2022]
Abstract
Branching morphogenesis drives the formation of epithelial organs including the mammary gland, lung, kidney, salivary gland and prostate. Branching at the cellular level also drives development of the nervous and vascular systems. A variety of signaling pathways are orchestrated together to establish the pattern of these branched organs. The phosphoinositide 3-kinase (PI3K) signaling network is of particular interest because of the diverse outcomes it generates, including proliferation, motility, growth, survival and cell death. Here, we focus on the role of the PI3K pathway in the development of branched tissues. Cultured cells, explants and transgenic mice have revealed that the PI3K pathway is critical for the regulation of cell proliferation, apoptosis and motility during branching of tissues.
Collapse
Affiliation(s)
- Wenting Zhu
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | | |
Collapse
|
137
|
Hsu J, Di Pasquale G, Harunaga J, Onodera T, Hoffman M, Chiorini J, Yamada K. Viral gene transfer to developing mouse salivary glands. J Dent Res 2012; 91:197-202. [PMID: 22095070 PMCID: PMC3261122 DOI: 10.1177/0022034511429346] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 09/30/2011] [Accepted: 10/03/2011] [Indexed: 11/16/2022] Open
Abstract
Branching morphogenesis is essential for the formation of salivary glands, kidneys, lungs, and many other organs during development, but the mechanisms underlying this process are not adequately understood. Microarray and other gene expression methods have been powerful approaches for identifying candidate genes that potentially regulate branching morphogenesis. However, functional validation of the proposed roles for these genes has been severely hampered by the absence of efficient techniques to genetically manipulate cells within embryonic organs. Using ex vivo cultured embryonic mouse submandibular glands (SMGs) as models to study branching morphogenesis, we have identified new vectors for viral gene transfer with high efficiency and cell-type specificity to developing SMGs. We screened adenovirus, lentivirus, and 11 types of adeno-associated viruses (AAV) for their ability to transduce embryonic day 12 or 13 SMGs. We identified two AAV types, AAV2 and bovine AAV (BAAV), that are selective in targeting expression differentially to SMG epithelial and mesenchymal cell populations, respectively. Transduction of SMG epithelia with self-complementary (sc) AAV2 expressing fibroblast growth factor 7 (Fgf7) supported gland survival and enhanced SMG branching morphogenesis. Our findings represent, to our knowledge, the first successful selective gene targeting to epithelial vs. mesenchymal cells in an organ undergoing branching morphogenesis.
Collapse
Affiliation(s)
- J.C. Hsu
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Building 30, Room 426, 30 Convent Drive, MSC 4370, Bethesda, MD 20892-4370, USA
| | - G. Di Pasquale
- Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Building 30, Room 426, 30 Convent Drive, MSC 4370, Bethesda, MD 20892-4370, USA
| | - J.S. Harunaga
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Building 30, Room 426, 30 Convent Drive, MSC 4370, Bethesda, MD 20892-4370, USA
| | - T. Onodera
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Building 30, Room 426, 30 Convent Drive, MSC 4370, Bethesda, MD 20892-4370, USA
| | - M.P. Hoffman
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Building 30, Room 426, 30 Convent Drive, MSC 4370, Bethesda, MD 20892-4370, USA
| | - J.A. Chiorini
- Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Building 30, Room 426, 30 Convent Drive, MSC 4370, Bethesda, MD 20892-4370, USA
| | - K.M. Yamada
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Building 30, Room 426, 30 Convent Drive, MSC 4370, Bethesda, MD 20892-4370, USA
| |
Collapse
|
138
|
Rebustini IT, Hayashi T, Reynolds AD, Dillard ML, Carpenter EM, Hoffman MP. miR-200c regulates FGFR-dependent epithelial proliferation via Vldlr during submandibular gland branching morphogenesis. Development 2011; 139:191-202. [PMID: 22115756 DOI: 10.1242/dev.070151] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The regulation of epithelial proliferation during organ morphogenesis is crucial for normal development, as dysregulation is associated with tumor formation. Non-coding microRNAs (miRNAs), such as miR-200c, are post-transcriptional regulators of genes involved in cancer. However, the role of miR-200c during normal development is unknown. We screened miRNAs expressed in the mouse developing submandibular gland (SMG) and found that miR-200c accumulates in the epithelial end buds. Using both loss- and gain-of-function, we demonstrated that miR-200c reduces epithelial proliferation during SMG morphogenesis. To identify the mechanism, we predicted miR-200c target genes and confirmed their expression during SMG development. We discovered that miR-200c targets the very low density lipoprotein receptor (Vldlr) and its ligand reelin, which unexpectedly regulate FGFR-dependent epithelial proliferation. Thus, we demonstrate that miR-200c influences FGFR-mediated epithelial proliferation during branching morphogenesis via a Vldlr-dependent mechanism. miR-200c and Vldlr may be novel targets for controlling epithelial morphogenesis during glandular repair or regeneration.
Collapse
Affiliation(s)
- Ivan T Rebustini
- Matrix and Morphogenesis Section, Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
139
|
Tsau C, Ito M, Gromova A, Hoffman MP, Meech R, Makarenkova HP. Barx2 and Fgf10 regulate ocular glands branching morphogenesis by controlling extracellular matrix remodeling. Development 2011; 138:3307-17. [PMID: 21750040 DOI: 10.1242/dev.066241] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The lacrimal gland (LG) develops through branching morphogenesis and produces secretions, including tears, that lubricate and protect the ocular surface. Despite the prevalence of LG disorders such as dry eye, relatively little is known about the regulation of LG development. In this study, we show that the homeobox transcription factor Barx2 is highly expressed in conjunctival epithelium, eyelids and ocular [lacrimal, harderian (HG), and meibomian (MG)] glands and is necessary for normal ocular gland and eyelid development. Barx2(-/-) mice show defective LG morphogenesis, absence of the HG, and defects in MG and eyelid fusion. Ex vivo antisense assays confirm the requirement for Barx2 in LG bud elongation and branching. Gene expression profiles reveal decreased expression of several adhesion and matrix remodeling molecules in Barx2(-/-) LGs. In culture, Barx2 regulates expression of matrix metalloproteinases (MMPs) and epithelial cell migration through the extracellular matrix. Fibroblast growth factors are crucial regulators of LG development and we show that Barx2 is required for Fgf10-induced LG bud elongation and that both Barx2 and Fgf10 cooperate in the regulation of MMPs. Together, these data suggest a mechanism for the effects of loss of Barx2 on ocular gland development. Intriguingly, salivary glands that also express a high level of Barx2 develop normally in Barx2(-/-) mice and do not show altered levels of MMPs. Thus, the function of Barx2 is specific to ocular gland development. Based on our data, we propose a functional network involving Barx2, Fgf10 and MMPs that plays an essential role in regulating branching morphogenesis of the ocular glands.
Collapse
Affiliation(s)
- Cindy Tsau
- The Neurosciences Institute, San Diego, CA 92121, USA
| | | | | | | | | | | |
Collapse
|
140
|
Häärä O, Fujimori S, Schmidt-Ullrich R, Hartmann C, Thesleff I, Mikkola ML. Ectodysplasin and Wnt pathways are required for salivary gland branching morphogenesis. Development 2011; 138:2681-91. [PMID: 21652647 DOI: 10.1242/dev.057711] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The developing submandibular salivary gland (SMG) is a well-studied model for tissue interactions and branching morphogenesis. Its development shares similar features with other ectodermal appendages such as hair and tooth. The ectodysplasin (Eda) pathway is essential for the formation and function of several ectodermal organs. Mutations in the signaling components of the Eda pathway lead to a human syndrome known as hypohidrotic ectodermal dysplasia (HED), which is characterized by missing and malformed teeth, sparse hair and reduced sweating. Individuals with HED suffer also from dry mouth because of reduced saliva flow. In order to understand the underlying mechanism, we analyzed salivary gland development in mouse models with altered Eda pathway activities. We have found that Eda regulates growth and branching of the SMG via transcription factor NF-κB in the epithelium, and that the hedgehog pathway is an important mediator of Eda/NF-κB. We also sought to determine whether a similar reciprocal interplay between the Eda and Wnt/β-catenin pathways, which are known to operate in other skin appendages, functions in developing SMG. Surprisingly and unlike in developing hair follicles and teeth, canonical Wnt signaling activity did not colocalize with Edar/NF-κB in salivary gland epithelium. Instead, we observed high mesenchymal Wnt activity and show that ablation of mesenchymal Wnt signaling either in vitro or in vivo compromised branching morphogenesis. We also provide evidence suggesting that the effects of mesenchymal Wnt/β-catenin signaling are mediated, at least in part, through regulation of Eda expression.
Collapse
Affiliation(s)
- Otso Häärä
- Developmental Biology Program, Institute of Biotechnology, University of Helsinki, P.O.B. 56, 00014 Helsinki, Finland
| | | | | | | | | | | |
Collapse
|
141
|
Patel N, Sharpe PT, Miletich I. Coordination of epithelial branching and salivary gland lumen formation by Wnt and FGF signals. Dev Biol 2011; 358:156-67. [PMID: 21806977 DOI: 10.1016/j.ydbio.2011.07.023] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Revised: 07/01/2011] [Accepted: 07/15/2011] [Indexed: 10/18/2022]
Abstract
Branching morphogenesis is a molecularly conserved mechanism that is adopted by several organs, such as the lung, kidney, mammary gland and salivary gland, to maximize the surface area of a tissue within a small volume. Branching occurs through repetitive clefting and elongation of spherical epithelial structures, called endbuds, which invade the surrounding mesenchyme. In the salivary gland, lumen formation takes place alongside branching morphogenesis, but in a controlled manner, so that branching is active at the distal ends of epithelial branches while lumen formation initiates at the proximal ends, and spreads distally. We present here data showing that interaction between FGF signaling and the canonical (β-catenin dependent) and non-canonical branches of Wnt signaling coordinates these two processes. Using the Axin2(lacZ) reporter mice, we find Wnt/β-catenin signaling activity first in the mesenchyme and later, at the time of lumen formation, in the ductal epithelium. Gain and loss of function experiments reveal that this pathway exerts an inhibitory effect on salivary gland branching morphogenesis. We have found that endbuds remain devoid of Wnt/β-catenin signaling activity, a hallmark of ductal structures, through FGF-mediated inhibition of this pathway. Our data also show that FGF signaling has a major role in the control of lumen formation by preventing premature hollowing of epithelial endbuds and slowing down the canalization of presumptive ducts. Concomitantly, FGF signaling strongly represses the ductal marker Cp2l1, most likely via repression of Wnt5b and non-canonical Wnt signaling. Inhibition of canonical and non-canonical Wnt signaling in endbuds by FGF signaling occurs at least in part through sFRP1, a secreted inhibitor of Wnt signaling and downstream target of FGF signaling. Altogether, these findings point to a key function of FGF signaling in the maintenance of an undifferentiated state in endbud cells by inhibition of a ductal fate.
Collapse
Affiliation(s)
- Nisha Patel
- Department of Craniofacial Development, Dental Institute, King's College London, Guy's Hospital, London Bridge, London, UK
| | | | | |
Collapse
|
142
|
Reciprocal interactions of Fgf10/Fgfr2b modulate the mouse tongue epithelial differentiation. Cell Tissue Res 2011; 345:265-73. [DOI: 10.1007/s00441-011-1204-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2010] [Accepted: 05/30/2011] [Indexed: 11/25/2022]
|
143
|
Miyajima H, Matsumoto T, Sakai T, Yamaguchi S, An SH, Abe M, Wakisaka S, Lee KY, Egusa H, Imazato S. Hydrogel-based biomimetic environment for in vitro modulation of branching morphogenesis. Biomaterials 2011; 32:6754-63. [PMID: 21683999 DOI: 10.1016/j.biomaterials.2011.05.072] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 05/25/2011] [Indexed: 11/15/2022]
Abstract
The mechanical properties of the cellular microenvironment dramatically alter during tissue development and growth. Growing evidence suggests that physical microenvironments and mechanical stresses direct cell fate in developing tissue. However, how these physical cues affect the tissue morphogenesis remains a major unknown. We explain here that the physical properties of the cell and tissue microenvironment, biomimetically reproduced by using hydrogel, guide the tissue morphogenesis in the developmental submandibular gland (SMG). In particular, the softer gel enhances the bud expansion and cleft formation of SMG, whereas the stiffer gel attenuates them. These morphological changes in SMG tissue are led by soluble factors (FGF7/10) induction regulated by cell traction force derived from the tissue deformation. Our findings suggest that cells sense the mechanics of their surrounding environment and alter their properties for self-organization and the following tissue morphogenesis. Also, physically designed hydrogel material is a valuable tool for producing the biomimetic microenvironment to explore how physical cues affect tissue morphogenesis and to modulate tissue morphogenesis for in vitro tissue synthesis.
Collapse
Affiliation(s)
- Hiroyuki Miyajima
- Department of Biomaterials Science, Osaka University, 1-8 Yamada-Oka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
144
|
The impact of compositional topography of amniotic membrane scaffold on tissue morphogenesis of salivary gland. Biomaterials 2011; 32:4424-32. [PMID: 21439637 DOI: 10.1016/j.biomaterials.2011.02.057] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Accepted: 02/27/2011] [Indexed: 12/16/2022]
Abstract
Amniotic membrane (AM) has been widely used in the reconstruction of oral epithelial defects. However, whether it is also effective in facilitating tissue formation of salivary gland, an appendix of oral epithelia, has never been explored. To investigate the effects and the underlying mechanism of AM on salivary gland morphogenesis, murine fetal submandibular gland (SMG) explants were cultured on different preparations of AM scaffolds. It was found that, on AM stromal scaffold, SMG demonstrated well-developed branching morphogenesis. Nonetheless, on AM epithelial scaffold, SMG epithelial cell converted to a spindle-shape, lost intercellular connection, changed cytoskeletal organization, and exhibited scattering behaviors. Meanwhile, the integrity of SMG basement membrane was dismantled as well. However, when acellular AM epithelial scaffold was used, cultured SMG demonstrated organized morphology, indicating that AM epithelial component provided specific surface features for SMG morphogenesis. To further investigate AM scaffold morphogenetic effect, it was found hepatocyte growth factor (HGF), an epithelial scattering factor, was expressed abundantly in cultivated AM epithelia. After blocking HGF function of AM, cultured SMG regained branching activity, reorganized cell adhesion and subcellular organization, and reproduced basement membranes. Therefore, AM-derived bioactive factor profoundly influences cell behaviors and structure formation of SMG. Together, this study showed that compositional topography of AM scaffold is important in affecting SMG morphogenesis. By understanding the effects of AM scaffold on SMG morphogenesis, it provides important information for rationally designing and fabricating AM scaffold for salivary gland regeneration.
Collapse
|
145
|
Hayashi T, Koyama N, Azuma Y, Kashimata M. Mesenchymal miR-21 regulates branching morphogenesis in murine submandibular gland in vitro. Dev Biol 2011; 352:299-307. [PMID: 21295561 DOI: 10.1016/j.ydbio.2011.01.030] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 01/17/2011] [Accepted: 01/25/2011] [Indexed: 01/01/2023]
Abstract
Branching morphogenesis in murine submandibular glands (SMG) is regulated by growth factors, extracellular matrix (ECM) and many biological processes through interactions between the epithelium and the mesenchyme. MicroRNAs (miRNAs) are a set of small, non-protein-coding RNAs that regulate gene expression at the post-transcriptional level. We hypothesized that branching morphogenesis is partly regulated by miRNAs. Forty-four miRNAs and novel miRNA candidates were detected in SMG at embryonic day 13 by a cloning method combined with Argonaute-2 immunoprecipitation. MicroRNA21 (miR-21) expression in the mesenchyme was up-regulated and accelerated by epidermal growth factor, which is known to enhance branching morphogenesis in vitro. Down-regulation of miR-21 in the mesenchyme by locked nucleic acids was associated with a decrease in the number of epithelial buds. Relative quantification of candidates for target genes of miR-21 indicated that two messenger RNAs (for Reck and Pdcd4) were down-regulated in the mesenchyme, where miR-21 expression levels were up-regulated. These results suggest that branching morphogenesis is regulated by miR-21 through gene expression related to ECM degradation in the mesenchyme.
Collapse
Affiliation(s)
- Toru Hayashi
- Department of Pharmacology, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu 501–0296, Japan
| | | | | | | |
Collapse
|
146
|
Koyama N, Hayashi T, Kashimata M. Regulation of Branching Morphogenesis in Fetal Mouse Submandibular Gland by Signaling Pathways Activated by Growth Factors and α6 Integrin. J Oral Biosci 2011. [DOI: 10.1016/s1349-0079(11)80022-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
147
|
Terao F, Takahashi I, Mitani H, Haruyama N, Sasano Y, Suzuki O, Takano-Yamamoto T. Fibroblast growth factor 10 regulates Meckel's cartilage formation during early mandibular morphogenesis in rats. Dev Biol 2010; 350:337-47. [PMID: 21147086 DOI: 10.1016/j.ydbio.2010.11.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Revised: 10/18/2010] [Accepted: 11/29/2010] [Indexed: 12/16/2022]
Abstract
Fibroblast growth factors (FGF) are pluripotent growth factors that play pivotal roles in the development of various organs. During mandibular organogenesis, Meckel's cartilage, teeth, and mandibular bone differentiate under the control of various FGF. In the present study, we evaluated the role of FGF10 in rat mandibular chondrogenesis and morphogenesis using mandibular organ culture and mandibular cell micromass culture systems. The overexpression of Fgf10 induced by the electroporation of an FGF10 expression vector not only altered the size and shape of Meckel's cartilage, but also upregulated the expression of the cartilage characteristic genes Col2a1 and Sox9 in a mandibular organ culture system. Meckel's cartilage was deformed, and its size was increased when Fgf10 was overexpressed in the lateral area of the mandible. Meanwhile, no effect was found when Fgf10 was overexpressed in the medial portion. In the mandibular cell micromass culture, recombinant FGF10 treatment enhanced chondrogenic differentiation and endogenous ERK (extracellular signal-regulated kinase) phosphorylation in cells derived from the lateral area of the mandible. On the other hand, FGF10 did not have significant effects on mandibular cell proliferation. These results indicate that FGF10 regulates Meckel's cartilage formation during early mandibular morphogenesis by controlling the cell differentiation in the lateral area of the mandibular process in rats.
Collapse
Affiliation(s)
- Fumie Terao
- Division of Orthodontics and Dentofacial Orthopedics, Tohoku University Graduate School of Dentistry, Aoba-ku, Sendai, Japan
| | | | | | | | | | | | | |
Collapse
|
148
|
Okamoto K, Kikuchi-Handa T, Nogawa H. Evidence of interlobular repulsion during branching morphogenesis in mouse salivary glands. Dev Dyn 2010; 239:2208-18. [PMID: 20568245 DOI: 10.1002/dvdy.22354] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
We developed a culture method for detecting repulsion among epithelial lobules during branching morphogenesis in mouse submandibular glands. Three epithelia were placed at each vertex of an imaginary triangle apart but near enough to meet with one another if each of them expands radially during the culture period. No repulsion was observed following cultivation with neuregulin 1 and lysophosphatidic acid; the epithelia actively branched and nearly contacted one another in the triangle's center. In contrast, strong repulsion was observed among the epithelia cultured with fibroblast growth factor 1 (FGF1), which exhibited less branching and moved away from the center. The localization of DiI- (1,1', di-octadecyl-3,3,3',3',-tetramethylindo-carbocyanine perchlorate) and BrdU- (5-bromodeoxyuridine) labeled cells in the cultures exposed to FGF1 indicated that the cells were unable to move and proliferate in the center. SB431542, an inhibitor of transforming growth factor-beta (TGFbeta) signaling, was unable to abolish this repulsion, suggesting that TGFbetas will not probably act as repellants in this case.
Collapse
Affiliation(s)
- Katsuya Okamoto
- Department of Biology, Graduate School of Science, Chiba University, Chiba 263-8522, Japan
| | | | | |
Collapse
|
149
|
Larsen M, Yamada KM, Musselmann K. Systems analysis of salivary gland development and disease. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2010; 2:670-82. [PMID: 20890964 PMCID: PMC3398465 DOI: 10.1002/wsbm.94] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Branching morphogenesis is a crucial developmental process in which vertebrate organs generate extensive epithelial surface area while retaining a compact size. In the vertebrate submandibular salivary gland, branching morphogenesis is crucial for the generation of the large surface area necessary to produce sufficient saliva. However, in many salivary gland diseases, saliva-producing acinar cells are destroyed, resulting in dry mouth and secondary health conditions. Systems-based approaches can provide insights into understanding salivary gland development, function, and disease. The traditional approach to understanding these processes is the identification of molecular signals using reductionist approaches; we review current progress with such methods in understanding salivary gland development. Taking a more global approach, multiple groups are currently profiling the transcriptome, the proteome, and other 'omes' in both developing mouse tissues and in human patient samples. Computational methods have been successful in deciphering large data sets, and mathematical models are starting to make predictions regarding the contribution of molecules to the physical processes of morphogenesis and cellular function. A challenge for the future will be to establish comprehensive, publicly accessible salivary gland databases spanning the full range of genes and proteins; plans are underway to provide these resources to researchers in centralized repositories. The greatest challenge for the future will be to develop realistic models that integrate multiple types of data to both describe and predict embryonic development and disease pathogenesis.
Collapse
Affiliation(s)
- Melinda Larsen
- Department of Biological Sciences, University at Albany, State University of New York
| | - Kenneth M. Yamada
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health
| | - Kurt Musselmann
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health
| |
Collapse
|
150
|
Knox S, Lombaert I, Reed X, Vitale-Cross L, Gutkind J, Hoffman M. Parasympathetic innervation maintains epithelial progenitor cells during salivary organogenesis. Science 2010; 329:1645-7. [PMID: 20929848 PMCID: PMC3376907 DOI: 10.1126/science.1192046] [Citation(s) in RCA: 245] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The maintenance of a progenitor cell population as a reservoir of undifferentiated cells is required for organ development and regeneration. However, the mechanisms by which epithelial progenitor cells are maintained during organogenesis are poorly understood. We report that removal of the parasympathetic ganglion in mouse explant organ culture decreased the number and morphogenesis of keratin 5-positive epithelial progenitor cells. These effects were rescued with an acetylcholine analog. We demonstrate that acetylcholine signaling, via the muscarinic M1 receptor and epidermal growth factor receptor, increased epithelial morphogenesis and proliferation of the keratin 5-positive progenitor cells. Parasympathetic innervation maintained the epithelial progenitor cell population in an undifferentiated state, which was required for organogenesis. This mechanism for epithelial progenitor cell maintenance may be targeted for organ repair or regeneration.
Collapse
Affiliation(s)
| | | | - X. Reed
- Matrix and Morphogenesis Unit, LCDB
| | - L Vitale-Cross
- OPCB, NIDCR, NIH, 30 Convent Dr, Bethesda, MD 20892, USA
| | - J.S. Gutkind
- OPCB, NIDCR, NIH, 30 Convent Dr, Bethesda, MD 20892, USA
| | | |
Collapse
|