1
|
Edirisinghe O, Ternier G, Alraawi Z, Suresh Kumar TK. Decoding FGF/FGFR Signaling: Insights into Biological Functions and Disease Relevance. Biomolecules 2024; 14:1622. [PMID: 39766329 PMCID: PMC11726770 DOI: 10.3390/biom14121622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Fibroblast Growth Factors (FGFs) and their cognate receptors, FGFRs, play pivotal roles in a plethora of biological processes, including cell proliferation, differentiation, tissue repair, and metabolic homeostasis. This review provides a comprehensive overview of FGF-FGFR signaling pathways while highlighting their complex regulatory mechanisms and interconnections with other signaling networks. Further, we briefly discuss the FGFs involvement in developmental, metabolic, and housekeeping functions. By complementing current knowledge and emerging research, this review aims to enhance the understanding of FGF-FGFR-mediated signaling and its implications for health and disease, which will be crucial for therapeutic development against FGF-related pathological conditions.
Collapse
Affiliation(s)
- Oshadi Edirisinghe
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA;
| | - Gaëtane Ternier
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, USA; (G.T.); (Z.A.)
| | - Zeina Alraawi
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, USA; (G.T.); (Z.A.)
| | - Thallapuranam Krishnaswamy Suresh Kumar
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA;
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, USA; (G.T.); (Z.A.)
| |
Collapse
|
2
|
Patel VN, Aure MH, Choi SH, Ball JR, Lane ED, Wang Z, Xu Y, Zheng C, Liu X, Martin D, Pailin JY, Prochazkova M, Kulkarni AB, van Kuppevelt TH, Ambudkar IS, Liu J, Hoffman MP. Specific 3-O-sulfated heparan sulfate domains regulate salivary gland basement membrane metabolism and epithelial differentiation. Nat Commun 2024; 15:7584. [PMID: 39217171 PMCID: PMC11365954 DOI: 10.1038/s41467-024-51862-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024] Open
Abstract
Heparan sulfate (HS) regulation of FGFR function, which is essential for salivary gland (SG) development, is determined by the immense structural diversity of sulfated HS domains. 3-O-sulfotransferases generate highly 3-O-sulfated HS domains (3-O-HS), and Hs3st3a1 and Hs3st3b1 are enriched in myoepithelial cells (MECs) that produce basement membrane (BM) and are a growth factor signaling hub. Hs3st3a1;Hs3st3b1 double-knockout (DKO) mice generated to investigate 3-O-HS regulation of MEC function and growth factor signaling show loss of specific highly 3-O-HS and increased FGF/FGFR complex binding to HS. During development, this increases FGFR-, BM- and MEC-related gene expression, while in adult, it reduces MECs, increases BM and disrupts acinar polarity, resulting in salivary hypofunction. Defined 3-O-HS added to FGFR pulldown assays and primary organ cultures modulates FGFR signaling to regulate MEC BM synthesis, which is critical for secretory unit homeostasis and acinar function. Understanding how sulfated HS regulates development will inform the use of HS mimetics in organ regeneration.
Collapse
Affiliation(s)
- Vaishali N Patel
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, NIH, DHHS, Bethesda, MD, USA.
| | - Marit H Aure
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, NIH, DHHS, Bethesda, MD, USA
| | - Sophie H Choi
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, NIH, DHHS, Bethesda, MD, USA
| | - James R Ball
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, NIH, DHHS, Bethesda, MD, USA
| | - Ethan D Lane
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, NIH, DHHS, Bethesda, MD, USA
| | - Zhangjie Wang
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
- Glycan Therapeutics Corp, Raleigh, NC, USA
| | - Yongmei Xu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Changyu Zheng
- Translational Research Core, National Institute of Dental and Craniofacial Research, NIH, DHHS, Bethesda, MD, USA
| | - Xibao Liu
- Secretory Physiology Section, National Institute of Dental and Craniofacial Research, NIH, DHHS, Bethesda, MD, USA
| | - Daniel Martin
- NIDCD/NIDCR Genomics and Computational Biology Core, National Institute of Dental and Craniofacial Research, NIH, DHHS, Bethesda, MD, USA
| | - Jillian Y Pailin
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, NIH, DHHS, Bethesda, MD, USA
| | - Michaela Prochazkova
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, NIH, DHHS, Bethesda, MD, USA
| | - Ashok B Kulkarni
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, NIH, DHHS, Bethesda, MD, USA
| | - Toin H van Kuppevelt
- Department of Biochemistry, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Indu S Ambudkar
- Secretory Physiology Section, National Institute of Dental and Craniofacial Research, NIH, DHHS, Bethesda, MD, USA
| | - Jian Liu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Matthew P Hoffman
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, NIH, DHHS, Bethesda, MD, USA.
| |
Collapse
|
3
|
Sekiguchi R, Martin D, Doyle A, Wang S, Yamada K. Salivary Gland Tissue Recombination Can Modify Cell Fate. J Dent Res 2024; 103:755-764. [PMID: 38715201 PMCID: PMC11191754 DOI: 10.1177/00220345241247484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024] Open
Abstract
Although mesenchyme is essential for inducing the epithelium of ectodermal organs, its precise role in organ-specific epithelial fate determination remains poorly understood. To elucidate the roles of tissue interactions in cellular differentiation, we performed single-cell RNA sequencing and imaging analyses on recombined tissues, where mesenchyme and epithelium were switched ex vivo between two types of embryonic mouse salivary glands: the parotid gland (a serous gland) and the submandibular gland (a predominantly mucous gland). We found partial induction of molecules that define gland-specific acinar and myoepithelial cells in recombined salivary epithelium. The parotid epithelium recombined with submandibular mesenchyme began to express mucous acinar genes not intrinsic to the parotid gland. While myoepithelial cells do not normally line parotid acini, newly induced myoepithelial cells densely populated recombined parotid acini. However, mucous acinar and myoepithelial markers continued to be expressed in submandibular epithelial cells recombined with parotid mesenchyme. Consequently, some epithelial cells appeared to be plastic, such that their fate could still be modified in response to mesenchymal signaling, whereas other epithelial cells appeared to be already committed to a specific fate. We also discovered evidence for bidirectional induction: transcriptional changes were observed not only in the epithelium but also in the mesenchyme after heterotypic tissue recombination. For example, parotid epithelium induced the expression of muscle-related genes in submandibular fibroblasts that began to mimic parotid fibroblast gene expression. These studies provide the first comprehensive unbiased molecular characterization of tissue recombination approaches exploring the regulation of cell fate.
Collapse
Affiliation(s)
- R. Sekiguchi
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - D. Martin
- Genomics and Computational Biology Core, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - A.D. Doyle
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
- Imaging Core, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - S. Wang
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | | | - K.M. Yamada
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
4
|
Wang B, Li Z, An W, Fan G, Li D, Qin L. Duct ligation/de-ligation model: exploring mechanisms for salivary gland injury and regeneration. Front Cell Dev Biol 2024; 12:1399934. [PMID: 38983787 PMCID: PMC11231214 DOI: 10.3389/fcell.2024.1399934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/07/2024] [Indexed: 07/11/2024] Open
Abstract
Sialadenitis and sialadenitis-induced sialopathy are typically caused by obstruction of the salivary gland ducts. Atrophy of the salivary glands in experimental animals caused by duct ligation exhibits a histopathology similar to that of salivary gland sialadenitis. Therefore, a variety of duct ligation/de-ligation models have been commonly employed to study salivary gland injury and regeneration. Duct ligation is mainly characterised by apoptosis and activation of different signaling pathways in parenchymal cells, which eventually leads to gland atrophy and progressive dysfunction. By contrast, duct de-ligation can initiate the recovery of gland structure and function by regenerating the secretory tissue. This review summarizes the animal duct ligation/de-ligation models that have been used for the examination of pathological fundamentals in salivary disorders, in order to unravel the pathological changes and underlying mechanisms involved in salivary gland injury and regeneration. These experimental models have contributed to developing effective and curative strategies for gland dysfunction and providing plausible solutions for overcoming salivary disorders.
Collapse
Affiliation(s)
- Bin Wang
- Department of Head and Neck Oncology, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Zhilin Li
- Department of Head and Neck Oncology, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Wei An
- Department of Oral and Maxillofacial Surgery, Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, China
| | - Gaiping Fan
- Department of Head and Neck Oncology, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Dezhi Li
- Department of Head and Neck Oncology, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
- Department of Head and Neck Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lizheng Qin
- Department of Oral and Maxillofacial and Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Tanaka J, Miura A, Shimamura Y, Hwang Y, Shimizu D, Kondo Y, Sawada A, Sarmah H, Ninish Z, Mishima K, Mori M. Generation of salivary glands derived from pluripotent stem cells via conditional blastocyst complementation. Cell Rep 2024; 43:114340. [PMID: 38865239 PMCID: PMC11580835 DOI: 10.1016/j.celrep.2024.114340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/25/2024] [Accepted: 05/23/2024] [Indexed: 06/14/2024] Open
Abstract
Whole salivary gland generation and transplantation offer potential therapies for salivary gland dysfunction. However, the specific lineage required to engineer complete salivary glands has remained elusive. In this study, we identify the Foxa2 lineage as a critical lineage for salivary gland development through conditional blastocyst complementation (CBC). Foxa2 lineage marking begins at the boundary between the endodermal and ectodermal regions of the oral epithelium before the formation of the primordial salivary gland, thereby labeling the entire gland. Ablation of Fgfr2 within the Foxa2 lineage in mice leads to salivary gland agenesis. We reversed this phenotype by injecting donor pluripotent stem cells into the mouse blastocysts, resulting in mice that survived to adulthood with salivary glands of normal size, comparable to those of their littermate controls. These findings demonstrate that CBC-based salivary gland regeneration serves as a foundational experimental approach for future advanced cell-based therapies.
Collapse
Affiliation(s)
- Junichi Tanaka
- Columbia Center for Human Development and Division of Pulmonary, Allergy, Critical Care, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA; Division of Pathology, Department of Oral Diagnostic Sciences, Showa University School of Dentistry, Tokyo 142-8555, Japan.
| | - Akihiro Miura
- Columbia Center for Human Development and Division of Pulmonary, Allergy, Critical Care, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Yuko Shimamura
- Columbia Center for Human Development and Division of Pulmonary, Allergy, Critical Care, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Youngmin Hwang
- Columbia Center for Human Development and Division of Pulmonary, Allergy, Critical Care, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Dai Shimizu
- Columbia Center for Human Development and Division of Pulmonary, Allergy, Critical Care, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Yuri Kondo
- Columbia Center for Human Development and Division of Pulmonary, Allergy, Critical Care, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Anri Sawada
- Columbia Center for Human Development and Division of Pulmonary, Allergy, Critical Care, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Hemanta Sarmah
- Columbia Stem Cell Initiative, Stem Cell Core, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Zurab Ninish
- Columbia Center for Human Development and Division of Pulmonary, Allergy, Critical Care, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Kenji Mishima
- Division of Pathology, Department of Oral Diagnostic Sciences, Showa University School of Dentistry, Tokyo 142-8555, Japan
| | - Munemasa Mori
- Columbia Center for Human Development and Division of Pulmonary, Allergy, Critical Care, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
6
|
Morales EA, Wang S. Salivary gland developmental mechanics. Curr Top Dev Biol 2024; 160:1-30. [PMID: 38937029 DOI: 10.1016/bs.ctdb.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
The salivary gland undergoes branching morphogenesis to elaborate into a tree-like structure with numerous saliva-secreting acinar units, all joined by a hierarchical ductal system. The expansive epithelial surface generated by branching morphogenesis serves as the structural basis for the efficient production and delivery of saliva. Here, we elucidate the process of salivary gland morphogenesis, emphasizing the role of mechanics. Structurally, the developing salivary gland is characterized by a stratified epithelium tightly encased by the basement membrane, which is in turn surrounded by a mesenchyme consisting of a dense network of interstitial matrix and mesenchymal cells. Diverse cell types and extracellular matrices bestow this developing organ with organized, yet spatially varied mechanical properties. For instance, the surface epithelial sheet of the bud is highly fluidic due to its high cell motility and weak cell-cell adhesion, rendering it highly pliable. In contrast, the inner core of the bud is more rigid, characterized by reduced cell motility and strong cell-cell adhesion, which likely provide structural support for the tissue. The interactions between the surface epithelial sheet and the inner core give rise to budding morphogenesis. Furthermore, the basement membrane and the mesenchyme offer mechanical constraints that could play a pivotal role in determining the higher-order architecture of a fully mature salivary gland.
Collapse
Affiliation(s)
- E Angelo Morales
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, United States
| | - Shaohe Wang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, United States.
| |
Collapse
|
7
|
Klangprapan J, Souza GR, Ferreira JN. Bioprinting salivary gland models and their regenerative applications. BDJ Open 2024; 10:39. [PMID: 38816372 PMCID: PMC11139920 DOI: 10.1038/s41405-024-00219-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 06/01/2024] Open
Abstract
OBJECTIVE Salivary gland (SG) hypofunction is a common clinical condition arising from radiotherapy to suppress head and neck cancers. The radiation often destroys the SG secretory acini, and glands are left with limited regenerative potential. Due to the complex architecture of SG acini and ducts, three-dimensional (3D) bioprinting platforms have emerged to spatially define these in vitro epithelial units and develop mini-organs or organoids for regeneration. Due to the limited body of evidence, this comprehensive review highlights the advantages and challenges of bioprinting platforms for SG regeneration. METHODS SG microtissue engineering strategies such as magnetic 3D bioassembly of cells and microfluidic coaxial 3D bioprinting of cell-laden microfibers and microtubes have been proposed to replace the damaged acinar units, avoid the use of xenogeneic matrices (like Matrigel), and restore salivary flow. RESULTS Replacing the SG damaged organ is challenging due to its complex architecture, which combines a ductal network with acinar epithelial units to facilitate a unidirectional flow of saliva. Our research group was the first to develop 3D bioassembly SG epithelial functional organoids with innervation to respond to both cholinergic and adrenergic stimulation. More recently, microtissue engineering using coaxial 3D bioprinting of hydrogel microfibers and microtubes could also supported the formation of viable epithelial units. Both bioprinting approaches could overcome the need for Matrigel by facilitating the assembly of adult stem cells, such as human dental pulp stem cells, and primary SG cells into micro-sized 3D constructs able to produce their own matrix and self-organize into micro-modular tissue clusters with lumenized areas. Furthermore, extracellular vesicle (EV) therapies from organoid-derived secretome were also designed and validated ex vivo for SG regeneration after radiation damage. CONCLUSION Magnetic 3D bioassembly and microfluidic coaxial bioprinting platforms have the potential to create SG mini-organs for regenerative applications via organoid transplantation or organoid-derived EV therapies.
Collapse
Affiliation(s)
- Jutapak Klangprapan
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Faculty of Dentistry, Chulalongkorn University, 34 Henri-Dunant Road, Pathumwan, Bangkok, 10330, Thailand
| | - Glauco R Souza
- Greiner Bio-one North America Inc., 4238 Capital Drive, Monroe, NC, 28110, USA
| | - João N Ferreira
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Faculty of Dentistry, Chulalongkorn University, 34 Henri-Dunant Road, Pathumwan, Bangkok, 10330, Thailand.
| |
Collapse
|
8
|
Wrynn T, Min S, Horeth E, Osinski J, Sinha S, Romano RA. ΔNp63 regulates Sfrp1 expression to direct salivary gland branching morphogenesis. PLoS One 2024; 19:e0301082. [PMID: 38722977 PMCID: PMC11081224 DOI: 10.1371/journal.pone.0301082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 03/08/2024] [Indexed: 05/13/2024] Open
Abstract
Branching morphogenesis is a complex process shared by many organs including the lungs, kidney, prostate, as well as several exocrine organs including the salivary, mammary and lacrimal glands. This critical developmental program ensures the expansion of an organ's surface area thereby maximizing processes of cellular secretion or absorption. It is guided by reciprocal signaling from the epithelial and mesenchymal cells. While signaling pathways driving salivary gland branching morphogenesis have been relatively well-studied, our understanding of the underlying transcriptional regulatory mechanisms directing this program, is limited. Here, we performed in vivo and ex vivo studies of the embryonic mouse submandibular gland to determine the function of the transcription factor ΔNp63, in directing branching morphogenesis. Our studies show that loss of ΔNp63 results in alterations in the differentiation program of the ductal cells which is accompanied by a dramatic reduction in branching morphogenesis that is mediated by dysregulation of WNT signaling. We show that ΔNp63 modulates WNT signaling to promote branching morphogenesis by directly regulating Sfrp1 expression. Collectively, our findings have revealed a novel role for ΔNp63 in the regulation of this critical process and offers a better understanding of the transcriptional networks involved in branching morphogenesis.
Collapse
Affiliation(s)
- Theresa Wrynn
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Sangwon Min
- Department of Stem Cell and Regenerative Biology, Faculty of Arts and Sciences, Harvard University, Cambridge, Massachusetts, United States of America
| | - Erich Horeth
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Jason Osinski
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Satrajit Sinha
- Department of Biochemistry, School of Dental Medicine, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Rose-Anne Romano
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, School of Dental Medicine, State University of New York at Buffalo, Buffalo, New York, United States of America
| |
Collapse
|
9
|
Gong J, Mita AC, Wei Z, Cheng HH, Mitchell EP, Wright JJ, Ivy SP, Wang V, Gray RC, McShane LM, Rubinstein LV, Patton DR, Williams PM, Hamilton SR, Tricoli JV, Conley BA, Arteaga CL, Harris LN, O’Dwyer PJ, Chen AP, Flaherty KT. Phase II Study of Erdafitinib in Patients With Tumors With Fibroblast Growth Factor Receptor Mutations or Fusions: Results From the NCI-MATCH ECOG-ACRIN Trial (EAY131) Subprotocol K2. JCO Precis Oncol 2024; 8:e2300407. [PMID: 38603650 PMCID: PMC11623915 DOI: 10.1200/po.23.00407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 11/14/2023] [Accepted: 02/28/2024] [Indexed: 04/13/2024] Open
Abstract
PURPOSE Subprotocol K2 (EAY131-K2) of the NCI-MATCH platform trial was an open-label, single-arm, phase II study designed to evaluate the antitumor efficacy of the oral FGFR1-4 inhibitor, erdafitinib, in patients with tumors harboring FGFR1-4 mutations or fusions. METHODS Central confirmation of tumor FGFR1-4 mutations or fusions was required for outcome analysis. Patients with urothelial carcinoma were excluded. Enrolled subjects received oral erdafitinib at a starting dose of 8 mg daily continuously until intolerable toxicity or disease progression. The primary end point was objective response rate (ORR) with key secondary end points of safety, progression-free survival (PFS), and overall survival (OS). RESULTS Thirty-five patients were enrolled, and 25 patients were included in the primary efficacy analysis as prespecified in the protocol. The median age was 61 years, and 52% of subjects had received ≥3 previous lines of therapy. The confirmed ORR was 16% (4 of 25 [90% CI, 5.7 to 33.0], P = .034 against the null rate of 5%). An additional seven patients experienced stable disease as best-confirmed response. Four patients had a prolonged PFS including two with recurrent WHO grade IV, IDH1-/2-wildtype glioblastoma. The median PFS and OS were 3.6 months and 11.0 months, respectively. Erdafitinib was manageable with no new safety signals. CONCLUSION This study met its primary end point in patients with several pretreated solid tumor types harboring FGFR1-3 mutations or fusions. These findings support advancement of erdafitinib for patients with fibroblast growth factor receptor-altered tumors outside of currently approved indications in a potentially tumor-agnostic manner.
Collapse
Affiliation(s)
- Jun Gong
- Cedars-Sinai Medical Center, Los Angeles, CA
| | | | - Zihan Wei
- Dana Farber Cancer Institute – ECOG-ACRIN Biostatistics Center, Boston, MA
| | | | - Edith P. Mitchell
- Sidney Kimmel Cancer Center at Thomas Jefferson University, Philadelphia, PA
| | - John J. Wright
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - S. Percy Ivy
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Victoria Wang
- Dana Farber Cancer Institute – ECOG-ACRIN Biostatistics Center, Boston, MA
| | - Robert C. Gray
- Dana Farber Cancer Institute – ECOG-ACRIN Biostatistics Center, Boston, MA
| | - Lisa M. McShane
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Larry V. Rubinstein
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - David R. Patton
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | | | | | - James V. Tricoli
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Barbara A. Conley
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | | | - Lyndsay N. Harris
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | | | - Alice P. Chen
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | | |
Collapse
|
10
|
Iwata D, Kometani-Gunjigake K, Nakao-Kuroishi K, Mizuhara M, Nakatomi M, Moriyama K, Ono K, Kawamoto T. Ser252Trp mutation in fibroblast growth factor receptor 2 promotes branching morphogenesis in mouse salivary glands. J Oral Biosci 2024; 66:90-97. [PMID: 38246420 DOI: 10.1016/j.job.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/23/2024]
Abstract
OBJECTIVES The purpose of this study was to perform morphological and immunohistochemical (IHC) analysis of the submandibular glands (SMGs) in early development in Apert syndrome model mice (Ap mice). METHODS ACTB-Cre homozygous mice were mated with fibroblast growth factor receptor 2 (Fgfr2+/Neo-S252W) mice; ACTB-Cre heterozygous mice (ACTB-Cre mice) at embryonic day (E) 13.5 served as the control group, and Fgfr2+/S252W mice (Ap mice) served as the experimental group. Hematoxylin and eosin (H&E) staining was performed on SMGs; Total SMG area and epithelial area were determined, and the epithelial occupancy ratio was calculated. Immunostaining was performed to assess the localization of FGF signaling-related proteins. Next, bromodeoxyuridine (BrdU)-positive cells were evaluated to assess cell proliferation. Finally, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining was performed to assess apoptosis in SMGs. RESULTS The epithelial occupancy ratio was significantly higher in SMGs of Ap mice compared with that in SMGs of controls. FGF7 and bone morphogenetic protein 4 (BMP4) exhibited different localizations in SMGs of Ap mice compared with SMGs of controls. Cell proliferation was higher in SMGs of Ap mice compared with that of controls; however, apoptosis did not different significantly between the two groups. CONCLUSION Our results suggest that enhanced FGF signaling conferred by missense mutations in FGFR2 promotes branching morphogenesis in SMGs of Ap mice.
Collapse
Affiliation(s)
- Daiki Iwata
- Division of Orofacial Functions and Orthodontics, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyusyu, Fukuoka, 803-8580, Japan
| | - Kaori Kometani-Gunjigake
- Division of Orofacial Functions and Orthodontics, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyusyu, Fukuoka, 803-8580, Japan
| | - Kayoko Nakao-Kuroishi
- Division of Orofacial Functions and Orthodontics, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyusyu, Fukuoka, 803-8580, Japan
| | - Masahiro Mizuhara
- Division of Orofacial Functions and Orthodontics, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyusyu, Fukuoka, 803-8580, Japan
| | - Mitsushiro Nakatomi
- Department of Human, Information and Life Sciences, School of Health Sciences, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyusyu, Fukuoka, 807-8580, Japan
| | - Keiji Moriyama
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Kentaro Ono
- Division of Physiology, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyusyu, Fukuoka, 803-8580, Japan
| | - Tatsuo Kawamoto
- Division of Orofacial Functions and Orthodontics, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyusyu, Fukuoka, 803-8580, Japan.
| |
Collapse
|
11
|
Rose SC, Larsen M, Xie Y, Sharfstein ST. Salivary Gland Bioengineering. Bioengineering (Basel) 2023; 11:28. [PMID: 38247905 PMCID: PMC10813147 DOI: 10.3390/bioengineering11010028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/19/2023] [Accepted: 11/30/2023] [Indexed: 01/23/2024] Open
Abstract
Salivary gland dysfunction affects millions globally, and tissue engineering may provide a promising therapeutic avenue. This review delves into the current state of salivary gland tissue engineering research, starting with a study of normal salivary gland development and function. It discusses the impact of fibrosis and cellular senescence on salivary gland pathologies. A diverse range of cells suitable for tissue engineering including cell lines, primary salivary gland cells, and stem cells are examined. Moreover, the paper explores various supportive biomaterials and scaffold fabrication methodologies that enhance salivary gland cell survival, differentiation, and engraftment. Innovative engineering strategies for the improvement of vascularization, innervation, and engraftment of engineered salivary gland tissue, including bioprinting, microfluidic hydrogels, mesh electronics, and nanoparticles, are also evaluated. This review underscores the promising potential of this research field for the treatment of salivary gland dysfunction and suggests directions for future exploration.
Collapse
Affiliation(s)
- Stephen C. Rose
- Department of Nanoscale Science and Engineering, College of Nanotechnology, Science, and Engineering, University at Albany, SUNY, 257 Fuller Road, Albany, NY 12203, USA (Y.X.)
| | - Melinda Larsen
- Department of Biological Sciences and The RNA Institute, University at Albany, SUNY, 1400 Washington Ave., Albany, NY 12222, USA;
| | - Yubing Xie
- Department of Nanoscale Science and Engineering, College of Nanotechnology, Science, and Engineering, University at Albany, SUNY, 257 Fuller Road, Albany, NY 12203, USA (Y.X.)
| | - Susan T. Sharfstein
- Department of Nanoscale Science and Engineering, College of Nanotechnology, Science, and Engineering, University at Albany, SUNY, 257 Fuller Road, Albany, NY 12203, USA (Y.X.)
| |
Collapse
|
12
|
Tanaka J, Miura A, Shimamura Y, Hwang Y, Shimizu D, Kondo Y, Sawada A, Sarmah H, Ninish Z, Mishima K, Mori M. Generation of salivary glands derived from pluripotent stem cells via conditional blastocyst complementation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.13.566845. [PMID: 38014349 PMCID: PMC10680620 DOI: 10.1101/2023.11.13.566845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Various patients suffer from dry mouth due to salivary gland dysfunction. Whole salivary gland generation and transplantation is a potential therapy to resolve this issue. However, the lineage permissible to design the entire salivary gland generation has been enigmatic. Here, we discovered Foxa2 as a lineage critical for generating a salivary gland via conditional blastocyst complementation (CBC). Foxa2 linage, but not Shh nor Pitx2, initiated to label between the boundary region of the endodermal and the ectodermal oral mucosa before primordial salivary gland formation, resulting in marking the entire salivary gland. The salivary gland was agenesis by depleting Fgfr2 under the Foxa2 lineage in the mice. We rescued this phenotype by injecting donor pluripotent stem cells into the mouse blastocysts. Those mice survived until adulthood with normal salivary glands compatible in size compared with littermate controls. These results indicated that CBC-based salivary gland generation is promising for next-generation cell-based therapy.
Collapse
|
13
|
McKendrick JG, Jones GR, Elder SS, Watson E, T'Jonck W, Mercer E, Magalhaes MS, Rocchi C, Hegarty LM, Johnson AL, Schneider C, Becher B, Pridans C, Mabbott N, Liu Z, Ginhoux F, Bajenoff M, Gentek R, Bain CC, Emmerson E. CSF1R-dependent macrophages in the salivary gland are essential for epithelial regeneration after radiation-induced injury. Sci Immunol 2023; 8:eadd4374. [PMID: 37922341 DOI: 10.1126/sciimmunol.add4374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 10/03/2023] [Indexed: 11/05/2023]
Abstract
The salivary glands often become damaged in individuals receiving radiotherapy for head and neck cancer, resulting in chronic dry mouth. This leads to detrimental effects on their health and quality of life, for which there is no regenerative therapy. Macrophages are the predominant immune cell in the salivary glands and are attractive therapeutic targets due to their unrivaled capacity to drive tissue repair. Yet, the nature and role of macrophages in salivary gland homeostasis and how they may contribute to tissue repair after injury are not well understood. Here, we show that at least two phenotypically and transcriptionally distinct CX3CR1+ macrophage populations are present in the adult salivary gland, which occupy anatomically distinct niches. CD11c+CD206-CD163- macrophages typically associate with gland epithelium, whereas CD11c-CD206+CD163+ macrophages associate with blood vessels and nerves. Using a suite of complementary fate mapping systems, we show that there are highly dynamic changes in the ontogeny and composition of salivary gland macrophages with age. Using an in vivo model of radiation-induced salivary gland injury combined with genetic or antibody-mediated depletion of macrophages, we demonstrate an essential role for macrophages in clearance of cells with DNA damage. Furthermore, we show that epithelial-associated macrophages are indispensable for effective tissue repair and gland function after radiation-induced injury, with their depletion resulting in reduced saliva production. Our data, therefore, provide a strong case for exploring the therapeutic potential of manipulating macrophages to promote tissue repair and thus minimize salivary gland dysfunction after radiotherapy.
Collapse
Affiliation(s)
- John G McKendrick
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Gareth-Rhys Jones
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Sonia S Elder
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Erin Watson
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Wouter T'Jonck
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Ella Mercer
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Marlene S Magalhaes
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Cecilia Rocchi
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Lizi M Hegarty
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Amanda L Johnson
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | | | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Clare Pridans
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Neil Mabbott
- Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Zhaoyuan Liu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Florent Ginhoux
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore 138648, Singapore
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Marc Bajenoff
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, INSERM, U1104, CNRS UMR7280, Marseille 13288, France
| | - Rebecca Gentek
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Calum C Bain
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Elaine Emmerson
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| |
Collapse
|
14
|
Aure MH, Symonds JM, Villapudua CU, Dodge JT, Werner S, Knosp WM, Hoffman MP. FGFR2 is essential for salivary gland duct homeostasis and MAPK-dependent seromucous acinar cell differentiation. Nat Commun 2023; 14:6485. [PMID: 37838739 PMCID: PMC10576811 DOI: 10.1038/s41467-023-42243-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 10/04/2023] [Indexed: 10/16/2023] Open
Abstract
Exocrine acinar cells in salivary glands (SG) are critical for oral health and loss of functional acinar cells is a major clinical challenge. Fibroblast growth factor receptors (FGFR) are essential for early development of multiple organs, including SG. However, the role of FGFR signaling in specific populations later in development and during acinar differentiation are unknown. Here, we use scRNAseq and conditional deletion of murine FGFRs in vivo to identify essential roles for FGFRs in craniofacial, early SG development and progenitor function during duct homeostasis. Importantly, we also discover that FGFR2 via MAPK signaling is critical for seromucous acinar differentiation and secretory gene expression, while FGFR1 is dispensable. We show that FGF7, expressed by myoepithelial cells (MEC), activates the FGFR2-dependent seromucous transcriptional program. Here, we propose a model where MEC-derived FGF7 drives seromucous acinar differentiation, providing a rationale for targeting FGFR2 signaling in regenerative therapies to restore acinar function.
Collapse
Affiliation(s)
- Marit H Aure
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA.
| | - Jennifer M Symonds
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Carlos U Villapudua
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Joshua T Dodge
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Sabine Werner
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology (ETH), Zurich, Zurich, Switzerland
| | - Wendy M Knosp
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Matthew P Hoffman
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
15
|
Marinkovic M, Tran ON, Wang H, Abdul-Azees P, Dean DD, Chen XD, Yeh CK. Autologous mesenchymal stem cells offer a new paradigm for salivary gland regeneration. Int J Oral Sci 2023; 15:18. [PMID: 37165024 PMCID: PMC10172302 DOI: 10.1038/s41368-023-00224-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 02/20/2023] [Accepted: 03/29/2023] [Indexed: 05/12/2023] Open
Abstract
Salivary gland (SG) dysfunction, due to radiotherapy, disease, or aging, is a clinical manifestation that has the potential to cause severe oral and/or systemic diseases and compromise quality of life. Currently, the standard-of-care for this condition remains palliative. A variety of approaches have been employed to restore saliva production, but they have largely failed due to damage to both secretory cells and the extracellular matrix (niche). Transplantation of allogeneic cells from healthy donors has been suggested as a potential solution, but no definitive population of SG stem cells, capable of regenerating the gland, has been identified. Alternatively, mesenchymal stem cells (MSCs) are abundant, well characterized, and during SG development/homeostasis engage in signaling crosstalk with the SG epithelium. Further, the trans-differentiation potential of these cells and their ability to regenerate SG tissues have been demonstrated. However, recent findings suggest that the "immuno-privileged" status of allogeneic adult MSCs may not reflect their status post-transplantation. In contrast, autologous MSCs can be recovered from healthy tissues and do not present a challenge to the recipient's immune system. With recent advances in our ability to expand MSCs in vitro on tissue-specific matrices, autologous MSCs may offer a new therapeutic paradigm for restoration of SG function.
Collapse
Affiliation(s)
- Milos Marinkovic
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Research Service, South Texas Veterans Health Care System, San Antonio, TX, USA
| | - Olivia N Tran
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Hanzhou Wang
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Parveez Abdul-Azees
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Research Service, South Texas Veterans Health Care System, San Antonio, TX, USA
| | - David D Dean
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, TX, USA
| | - Xiao-Dong Chen
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
- Research Service, South Texas Veterans Health Care System, San Antonio, TX, USA.
- Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, TX, USA.
| | - Chih-Ko Yeh
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
- Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, TX, USA.
| |
Collapse
|
16
|
Gao J, Li A, Fujii S, Huang F, Nakatomi C, Nakamura I, Honda H, Kiyoshima T, Jimi E. p130Cas is required for androgen-dependent postnatal development regulation of submandibular glands. Sci Rep 2023; 13:5144. [PMID: 36991029 PMCID: PMC10060253 DOI: 10.1038/s41598-023-32390-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
Salivary glands develop through epithelial-mesenchymal interactions and are formed through repeated branching. The Crk-associated substrate protein (p130Cas) serves as an adapter that forms a complex with various proteins via integrin and growth factor signaling, with important regulatory roles in several essential cellular processes. We found that p130Cas is expressed in ductal epithelial cells of the submandibular gland (SMG). We generated epithelial tissue-specific p130Cas-deficient (p130CasΔepi-) mice and aimed to investigate the physiological role of p130Cas in the postnatal development of salivary glands. Histological analysis showed immature development of granular convoluted tubules (GCT) of the SMG in male p130CasΔepi- mice. Immunofluorescence staining showed that nuclear-localized androgen receptors (AR) were specifically decreased in GCT cells in p130CasΔepi- mice. Furthermore, epidermal growth factor-positive secretory granules contained in GCT cells were significantly reduced in p130CasΔepi- mice with downregulated AR signaling. GCTs lacking p130Cas showed reduced numbers and size of secretory granules, disrupted subcellular localization of the cis-Golgi matrix protein GM130, and sparse endoplasmic reticulum membranes in GCT cells. These results suggest that p130Cas plays a crucial role in androgen-dependent GCT development accompanied with ER-Golgi network formation in SMG by regulating the AR signaling.
Collapse
Affiliation(s)
- Jing Gao
- Laboratory of Molecular and Cellular Biochemistry, Division of Oral Biological Sciences, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Aonan Li
- Laboratory of Molecular and Cellular Biochemistry, Division of Oral Biological Sciences, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Shinsuke Fujii
- Laboratory of Oral Pathology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
- Dento-Craniofacial Development and Regeneration Research Center Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Fei Huang
- Laboratory of Molecular and Cellular Biochemistry, Division of Oral Biological Sciences, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Chihiro Nakatomi
- Division of Physiology, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, 803-8580, Japan
| | - Ichiro Nakamura
- Department of Rehabilitation, Yugawara Hospital, Japan Community Health Care Organization, 2-21-6 Chuo, Yugawara, Ashigara-shimo, Kanagawa, 259-0396, Japan
| | - Hiroaki Honda
- Field of Human Disease Models, Major in Advanced Life Sciences and Medicine, Institute of Laboratory Animals, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Tamotsu Kiyoshima
- Laboratory of Oral Pathology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Eijiro Jimi
- Laboratory of Molecular and Cellular Biochemistry, Division of Oral Biological Sciences, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
- Oral Health/Brain Health/Total Health Research Center, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
17
|
Chibly AM, Patel VN, Aure MH, Pasquale MC, Martin GE, Ghannam M, Andrade J, Denegre NG, Simpson C, Goldstein DP, Liu FF, Lombaert IMA, Hoffman MP. Neurotrophin signaling is a central mechanism of salivary dysfunction after irradiation that disrupts myoepithelial cells. NPJ Regen Med 2023; 8:17. [PMID: 36966175 PMCID: PMC10039923 DOI: 10.1038/s41536-023-00290-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 02/27/2023] [Indexed: 03/27/2023] Open
Abstract
The mechanisms that prevent regeneration of irradiated (IR) salivary glands remain elusive. Bulk RNAseq of IR versus non-IR human salivary glands showed that neurotrophin signaling is highly disrupted post-radiation. Neurotrophin receptors (NTRs) were significantly upregulated in myoepithelial cells (MECs) post-IR, and single cell RNAseq revealed that MECs pericytes, and duct cells are the main sources of neurotrophin ligands. Using two ex vivo models, we show that nerve growth factor (NGF) induces expression of MEC genes during development, and upregulation of NTRs in adult MECs is associated with stress-induced plasticity and morphological abnormalities in IR human glands. As MECs are epithelial progenitors after gland damage and are required for proper acinar cell contraction and secretion, we propose that MEC-specific upregulation of NTRs post-IR disrupts MEC differentiation and potentially impedes the ability of the gland to regenerate.
Collapse
Affiliation(s)
- Alejandro M Chibly
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Vaishali N Patel
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Marit H Aure
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mary C Pasquale
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Gemma E Martin
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mousa Ghannam
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Julianne Andrade
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Noah G Denegre
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Colleen Simpson
- Department of Otolaryngology-Head & Neck Surgery, Princess Margaret Cancer Center, Toronto, ON, M5G 2C4, Canada
| | - David P Goldstein
- Department of Otolaryngology-Head & Neck Surgery, Princess Margaret Cancer Center, Toronto, ON, M5G 2C4, Canada
| | - Fei-Fei Liu
- Department of Radiation Oncology, Princess Margaret Cancer Center, Toronto, ON, M5G 2M9, Canada
| | - Isabelle M A Lombaert
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
- Department of Biologic and Material Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI, 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Matthew P Hoffman
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
18
|
Ray AT, Soriano P. FGF signaling regulates salivary gland branching morphogenesis by modulating cell adhesion. Development 2023; 150:dev201293. [PMID: 36861436 PMCID: PMC10112918 DOI: 10.1242/dev.201293] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 02/22/2023] [Indexed: 03/03/2023]
Abstract
Loss of FGF signaling leads to defects in salivary gland branching, but the mechanisms underlying this phenotype remain largely unknown. We disrupted expression of Fgfr1 and Fgfr2 in salivary gland epithelial cells and found that both receptors function coordinately in regulating branching. Strikingly, branching morphogenesis in double knockouts is restored by Fgfr1 and Fgfr2 (Fgfr1/2) knock-in alleles incapable of engaging canonical RTK signaling, suggesting that additional FGF-dependent mechanisms play a role in salivary gland branching. Fgfr1/2 conditional null mutants showed defective cell-cell and cell-matrix adhesion, both of which have been shown to play instructive roles in salivary gland branching. Loss of FGF signaling led to disordered cell-basement membrane interactions in vivo as well as in organ culture. This was partially restored upon introducing Fgfr1/2 wild-type or signaling alleles that are incapable of eliciting canonical intracellular signaling. Together, our results identify non-canonical FGF signaling mechanisms that regulate branching morphogenesis through cell-adhesion processes.
Collapse
Affiliation(s)
- Ayan T. Ray
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Philippe Soriano
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
19
|
Sekiguchi R, Mehlferber M, Matsumoto K, Wang S. Efficient Gene Knockout in Salivary Gland Epithelial Explant Cultures. J Dent Res 2023; 102:197-206. [PMID: 36366748 PMCID: PMC9893391 DOI: 10.1177/00220345221128201] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We have developed methods to achieve efficient CRISPR-Cas9-mediated gene knockout in ex vivo mouse embryonic salivary epithelial explants. Salivary epithelial explants provide a valuable model for characterizing cell signaling, differentiation, and epithelial morphogenesis, but research has been limited by a paucity of efficient gene perturbation methods. Here, we demonstrate highly efficient gene perturbation by transient transduction of guide RNA-expressing lentiviruses into Cas9-expressing salivary epithelial buds isolated from Cas9 transgenic mice. We first show that salivary epithelial explants can be cultured in low-concentration, nonsolidified Matrigel suspensions in 96-well plates, which greatly increases sample throughput compared to conventional cultures embedded in solidified Matrigel. We further show that salivary epithelial explants can grow and branch with FGF7 alone, while supplementing with insulin, transferrin, and selenium (ITS) enhances growth and branching. We then describe an efficient workflow to produce experiment-ready, high-titer lentiviruses within 1 wk after molecular cloning. To track transduced cells, we designed the lentiviral vector to coexpress a nuclear fluorescent reporter with the guide RNA. We routinely achieved 80% transduction efficiency when antibiotic selection was used. Importantly, we detected robust loss of targeted protein products when testing 9 guide RNAs for 3 different genes. Moreover, targeting the β1 integrin gene (Itgb1) inhibited branching morphogenesis, which supports the importance of cell-matrix adhesion in driving branching morphogenesis. In summary, we have established a lentivirus-based method that can efficiently perturb genes of interest in salivary epithelial explants, which will greatly facilitate studies of specific gene functions using this system.
Collapse
Affiliation(s)
- R. Sekiguchi
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - M.M. Mehlferber
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
- Present address: Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
| | - K. Matsumoto
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - S. Wang
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
- Present address: 4D Cellular Physiology, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| |
Collapse
|
20
|
Chansaenroj A, Adine C, Charoenlappanit S, Roytrakul S, Sariya L, Osathanon T, Rungarunlert S, Urkasemsin G, Chaisuparat R, Yodmuang S, Souza GR, Ferreira JN. Magnetic bioassembly platforms towards the generation of extracellular vesicles from human salivary gland functional organoids for epithelial repair. Bioact Mater 2022; 18:151-163. [PMID: 35387159 PMCID: PMC8961305 DOI: 10.1016/j.bioactmat.2022.02.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 01/27/2022] [Accepted: 02/08/2022] [Indexed: 11/04/2022] Open
Abstract
Salivary glands (SG) are exocrine organs with secretory units commonly injured by radiotherapy. Bio-engineered organoids and extracellular vesicles (EV) are currently under investigation as potential strategies for SG repair. Herein, three-dimensional (3D) cultures of SG functional organoids (SGo) and human dental pulp stem cells (hDPSC) were generated by magnetic 3D bioassembly (M3DB) platforms. Fibroblast growth factor 10 (FGF10) was used to enrich the SGo in secretory epithelial units. After 11 culture days via M3DB, SGo displayed SG-specific acinar epithelial units with functional properties upon neurostimulation. To consistently develop 3D hDPSC in vitro, 3 culture days were sufficient to maintain hDPSC undifferentiated genotype and phenotype for EV generation. EV isolation was performed via sequential centrifugation of the conditioned media of hDPSC and SGo cultures. EV were characterized by nanoparticle tracking analysis, electron microscopy and immunoblotting. EV were in the exosome range for hDPSC (diameter: 88.03 ± 15.60 nm) and for SGo (123.15 ± 63.06 nm). Upon ex vivo administration, exosomes derived from SGo significantly stimulated epithelial growth (up to 60%), mitosis, epithelial progenitors and neuronal growth in injured SG; however, such biological effects were less distinctive with the ones derived from hDPSC. Next, these exosome biological effects were investigated by proteomic arrays. Mass spectrometry profiling of SGo exosomes predicted that cellular growth, development and signaling was due to known and undocumented molecular targets downstream of FGF10. Semaphorins were identified as one of the novel targets requiring further investigations. Thus, M3DB platforms can generate exosomes with potential to ameliorate SG epithelial damage.
Collapse
Affiliation(s)
- Ajjima Chansaenroj
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Christabella Adine
- Faculty of Dentistry, National University of Singapore, 119077, Singapore, Singapore
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, 119077, Singapore, Singapore
| | - Sawanya Charoenlappanit
- Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Sittiruk Roytrakul
- Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Ladawan Sariya
- The Monitoring and Surveillance Center for Zoonotic Diseases in Wildlife and Exotic Animals, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Thanaphum Osathanon
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Sasitorn Rungarunlert
- Department of Preclinical and Applied Animal Science, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Ganokon Urkasemsin
- Department of Preclinical and Applied Animal Science, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Risa Chaisuparat
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Supansa Yodmuang
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
- Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Glauco R. Souza
- University of Texas Health Sciences Center at Houston, Houston, TX, 77030, USA
- Nano3D Biosciences Inc., Houston, TX, 77030, USA
- Greiner Bio-One North America Inc, Monroe, NC, 28110, USA
| | - João N. Ferreira
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
- Faculty of Dentistry, National University of Singapore, 119077, Singapore, Singapore
| |
Collapse
|
21
|
May AJ, Mattingly AJ, Gaylord EA, Griffin N, Sudiwala S, Cruz-Pacheco N, Emmerson E, Mohabbat S, Nathan S, Sinada H, Lombaert IMA, Knox SM. Neuronal-epithelial cross-talk drives acinar specification via NRG1-ERBB3-mTORC2 signaling. Dev Cell 2022; 57:2550-2565.e5. [PMID: 36413949 PMCID: PMC9727910 DOI: 10.1016/j.devcel.2022.10.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 09/14/2022] [Accepted: 10/26/2022] [Indexed: 11/23/2022]
Abstract
Acinar cells are the principal secretory units of multiple exocrine organs. A single-cell, layered, lumenized acinus forms from a large cohort of epithelial progenitors that must initiate and coordinate three cellular programs of acinar specification, namely, lineage progression, secretion, and polarization. Despite this well-known outcome, the mechanism(s) that regulate these complex programs are unknown. Here, we demonstrate that neuronal-epithelial cross-talk drives acinar specification through neuregulin (NRG1)-ERBB3-mTORC2 signaling. Using single-cell and global RNA sequencing of developing murine salivary glands, we identified NRG1-ERBB3 to precisely overlap with acinar specification during gland development. Genetic deletion of Erbb3 prevented cell lineage progression and the establishment of lumenized, secretory acini. Conversely, NRG1 treatment of isolated epithelia was sufficient to recapitulate the development of secretory acini. Mechanistically, we found that NRG1-ERBB3 regulates each developmental program through an mTORC2 signaling pathway. Thus, we reveal that a neuronal-epithelial (NRG1/ERBB3/mTORC2) mechanism orchestrates the creation of functional acini.
Collapse
Affiliation(s)
- Alison J May
- Program in Craniofacial Biology, University of California, 513 Parnassus Avenue, San Francisco, CA 94143, USA; Department of Cell and Tissue Biology, University of California, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Aaron J Mattingly
- Program in Craniofacial Biology, University of California, 513 Parnassus Avenue, San Francisco, CA 94143, USA; Department of Cell and Tissue Biology, University of California, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Eliza A Gaylord
- Program in Craniofacial Biology, University of California, 513 Parnassus Avenue, San Francisco, CA 94143, USA; Department of Cell and Tissue Biology, University of California, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Nathan Griffin
- Program in Craniofacial Biology, University of California, 513 Parnassus Avenue, San Francisco, CA 94143, USA; Department of Cell and Tissue Biology, University of California, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Sonia Sudiwala
- Program in Craniofacial Biology, University of California, 513 Parnassus Avenue, San Francisco, CA 94143, USA; Department of Cell and Tissue Biology, University of California, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Noel Cruz-Pacheco
- Program in Craniofacial Biology, University of California, 513 Parnassus Avenue, San Francisco, CA 94143, USA; Department of Cell and Tissue Biology, University of California, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Elaine Emmerson
- Program in Craniofacial Biology, University of California, 513 Parnassus Avenue, San Francisco, CA 94143, USA; Department of Cell and Tissue Biology, University of California, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Seayar Mohabbat
- Program in Craniofacial Biology, University of California, 513 Parnassus Avenue, San Francisco, CA 94143, USA; Department of Cell and Tissue Biology, University of California, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Sara Nathan
- Program in Craniofacial Biology, University of California, 513 Parnassus Avenue, San Francisco, CA 94143, USA; Department of Cell and Tissue Biology, University of California, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Hanan Sinada
- Program in Craniofacial Biology, University of California, 513 Parnassus Avenue, San Francisco, CA 94143, USA; Department of Cell and Tissue Biology, University of California, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Isabelle M A Lombaert
- Biointerfaces Institute, University of Michigan, 2800 Plymouth Rd, Ann Arbor, MI 48109, USA; Department of Biologic and Materials Sciences & Prosthodontics, School of Dentistry, University of Michigan, 1011 N University Ave, Ann Arbor, MI 48109, USA.
| | - Sarah M Knox
- Program in Craniofacial Biology, University of California, 513 Parnassus Avenue, San Francisco, CA 94143, USA; Department of Cell and Tissue Biology, University of California, 513 Parnassus Avenue, San Francisco, CA 94143, USA.
| |
Collapse
|
22
|
Ehnes DD, Alghadeer A, Hanson-Drury S, Zhao YT, Tilmes G, Mathieu J, Ruohola-Baker H. Sci-Seq of Human Fetal Salivary Tissue Introduces Human Transcriptional Paradigms and a Novel Cell Population. FRONTIERS IN DENTAL MEDICINE 2022; 3:887057. [PMID: 36540608 PMCID: PMC9762771 DOI: 10.3389/fdmed.2022.887057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2023] Open
Abstract
Multiple pathologies and non-pathological factors can disrupt the function of the non-regenerative human salivary gland including cancer and cancer therapeutics, autoimmune diseases, infections, pharmaceutical side effects, and traumatic injury. Despite the wide range of pathologies, no therapeutic or regenerative approaches exist to address salivary gland loss, likely due to significant gaps in our understanding of salivary gland development. Moreover, identifying the tissue of origin when diagnosing salivary carcinomas requires an understanding of human fetal development. Using computational tools, we identify developmental branchpoints, a novel stem cell-like population, and key signaling pathways in the human developing salivary glands by analyzing our human fetal single-cell sequencing data. Trajectory and transcriptional analysis suggest that the earliest progenitors yield excretory duct and myoepithelial cells and a transitional population that will yield later ductal cell types. Importantly, this single-cell analysis revealed a previously undescribed population of stem cell-like cells that are derived from SD and expresses high levels of genes associated with stem cell-like function. We have observed these rare cells, not in a single niche location but dispersed within the developing duct at later developmental stages. Our studies introduce new human-specific developmental paradigms for the salivary gland and lay the groundwork for the development of translational human therapeutics.
Collapse
Affiliation(s)
- Devon Duron Ehnes
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cells and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
| | - Ammar Alghadeer
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cells and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
- Department of Biomedical Dental Sciences, College of Dentistry, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
| | - Sesha Hanson-Drury
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cells and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
- Department of Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA, United States
| | - Yan Ting Zhao
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cells and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
- Department of Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA, United States
| | - Gwen Tilmes
- Institute for Stem Cells and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
| | - Julie Mathieu
- Institute for Stem Cells and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
- Department of Comparative Medicine, University of Washington, Seattle, WA, United States
| | - Hannele Ruohola-Baker
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cells and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States
- Department of Biomedical Dental Sciences, College of Dentistry, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| |
Collapse
|
23
|
Xiaojie W, Banda J, Qi H, Chang AK, Bwalya C, Chao L, Li X. Scarless wound healing: Current insights from the perspectives of TGF-β, KGF-1, and KGF-2. Cytokine Growth Factor Rev 2022; 66:26-37. [PMID: 35690568 DOI: 10.1016/j.cytogfr.2022.03.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 03/22/2022] [Indexed: 11/03/2022]
Abstract
The process of wound healing involves a complex and vast interplay of growth factors and cytokines that coordinate the recruitment and interaction of various cell types. A series of events involving inflammation, proliferation, and remodeling eventually leads to the restoration of the damaged tissue. Abrogation in the regulation of these events has been shown to result in excessive scarring or non-healing wounds. While the process of wound healing is not fully elucidated, it has been documented that the early events of wound healing play a key role in the outcome of the wound. Furthermore, high levels of inflammation have been shown to lead to scarring. The regulation of these events may result in scarless wound healing, especially in adults. The inhibition of transforming growth factor-β (TGF-β) and the administration of keratinocyte growth factors (KGF), KGF-1 and KGF-2, has in recent years yielded positive results in the acceleration of wound closure and reduced scarring. Here, we encapsulate recent knowledge on the roles of TGF-β, KGF1, and KGF2 in wound healing and scar formation and highlight the areas that need further investigation. We also discuss potential future directions for the use of growth factors in wound management.
Collapse
Affiliation(s)
| | | | - Hui Qi
- Wenzhou Medical University, China
| | | | | | - Lu Chao
- Wenzhou Medical University, China
| | | |
Collapse
|
24
|
Retinoic acid and FGF10 promote the differentiation of pluripotent stem cells into salivary gland placodes. Stem Cell Res Ther 2022; 13:368. [PMID: 35902913 PMCID: PMC9330698 DOI: 10.1186/s13287-022-03033-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 07/05/2022] [Indexed: 11/10/2022] Open
Abstract
Background Salivary glands produce saliva that play essential roles in digestion and oral health. Derivation of salivary gland organoids from pluripotent stem cells (PSCs) provides a powerful platform to model the organogenesis processes during development. A few studies attempted to differentiate PSCs into salivary gland organoids. However, none of them could recapitulate the morphogenesis of the embryonic salivary glands, and most of the protocols involved complicated manufacturing processes. Methods To generate PSC-derived salivary gland placodes, the mouse embryonic stem cells were first differentiated into oral ectoderm by treatment with BMP4 on day 3. Retinoic acid and bFGF were then applied to the cultures from day 4 to day 6, followed by a 4-day treatment of FGF10. The PSC-derived salivary gland placodes on day 10 were transplanted to kidney capsules to determine the regenerative potential. Quantitative reverse transcriptase–polymerase chain reaction, immunofluorescence, and RNA-sequencing were performed to identify the PSC-derived SG placodes. Results We showed that step-wise treatment of retinoic acid and FGF10 promoted the differentiation of PSCs into salivary gland placodes, which can recapitulate the early morphogenetic events of their fetal counterparts, including the thickening, invagination, and then formed initial buds. The PSC-derived salivary gland placodes also differentiated into developing duct structures and could develop to striated and excretory ducts when transplanted in vivo. Conclusions The present study provided an easy and safe method to generate salivary gland placodes from PSCs, which offered possibilities for studying salivary gland development in vitro and developing new cell therapies. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03033-5.
Collapse
|
25
|
Chibly AM, Aure MH, Patel VN, Hoffman MP. Salivary gland function, development, and regeneration. Physiol Rev 2022; 102:1495-1552. [PMID: 35343828 PMCID: PMC9126227 DOI: 10.1152/physrev.00015.2021] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 11/27/2021] [Accepted: 03/17/2022] [Indexed: 02/08/2023] Open
Abstract
Salivary glands produce and secrete saliva, which is essential for maintaining oral health and overall health. Understanding both the unique structure and physiological function of salivary glands, as well as how they are affected by disease and injury, will direct the development of therapy to repair and regenerate them. Significant recent advances, particularly in the OMICS field, increase our understanding of how salivary glands develop at the cellular, molecular, and genetic levels: the signaling pathways involved, the dynamics of progenitor cell lineages in development, homeostasis, and regeneration, and the role of the extracellular matrix microenvironment. These provide a template for cell and gene therapies as well as bioengineering approaches to repair or regenerate salivary function.
Collapse
Affiliation(s)
- Alejandro M Chibly
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| | - Marit H Aure
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| | - Vaishali N Patel
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| | - Matthew P Hoffman
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
26
|
Piraino LR, Benoit DSW, DeLouise LA. Optimizing Soluble Cues for Salivary Gland Tissue Mimetics Using a Design of Experiments (DoE) Approach. Cells 2022; 11:1962. [PMID: 35741092 PMCID: PMC9222211 DOI: 10.3390/cells11121962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/14/2022] [Accepted: 06/14/2022] [Indexed: 02/01/2023] Open
Abstract
The development of therapies to prevent or treat salivary gland dysfunction has been limited by a lack of functional in vitro models. Specifically, critical markers of salivary gland secretory phenotype downregulate rapidly ex vivo. Here, we utilize a salivary gland tissue chip model to conduct a design of experiments (DoE) approach to test combinations of seven soluble cues that were previously shown to maintain or improve salivary gland cell function. This approach uses statistical techniques to improve efficiency and accuracy of combinations of factors. The DoE-designed culture conditions improve markers of salivary gland function. Data show that the EGFR inhibitor, EKI-785, maintains relative mRNA expression of Mist1, a key acinar cell transcription factor, while FGF10 and neurturin promote mRNA expression of Aqp5 and Tmem16a, channel proteins involved in secretion. Mist1 mRNA expression correlates with increased secretory function, including calcium signaling and mucin (PAS-AB) staining. Overall, this study demonstrates that media conditions can be efficiently optimized to support secretory function in vitro using a DoE approach.
Collapse
Affiliation(s)
- Lindsay R. Piraino
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, USA; (L.R.P.); (D.S.W.B.)
| | - Danielle S. W. Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, USA; (L.R.P.); (D.S.W.B.)
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
- Materials Science Program, University of Rochester, Rochester, NY 14627, USA
- Department of Chemical Engineering, University of Rochester, Rochester, NY 14627, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Lisa A. DeLouise
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, USA; (L.R.P.); (D.S.W.B.)
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Materials Science Program, University of Rochester, Rochester, NY 14627, USA
- Department of Dermatology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
27
|
Hajiabbas M, D'Agostino C, Simińska-Stanny J, Tran SD, Shavandi A, Delporte C. Bioengineering in salivary gland regeneration. J Biomed Sci 2022; 29:35. [PMID: 35668440 PMCID: PMC9172163 DOI: 10.1186/s12929-022-00819-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/26/2022] [Indexed: 11/16/2022] Open
Abstract
Salivary gland (SG) dysfunction impairs the life quality of many patients, such as patients with radiation therapy for head and neck cancer and patients with Sjögren’s syndrome. Multiple SG engineering strategies have been considered for SG regeneration, repair, or whole organ replacement. An in-depth understanding of the development and differentiation of epithelial stem and progenitor cells niche during SG branching morphogenesis and signaling pathways involved in cell–cell communication constitute a prerequisite to the development of suitable bioengineering solutions. This review summarizes the essential bioengineering features to be considered to fabricate an engineered functional SG model using various cell types, biomaterials, active agents, and matrix fabrication methods. Furthermore, recent innovative and promising approaches to engineering SG models are described. Finally, this review discusses the different challenges and future perspectives in SG bioengineering.
Collapse
Affiliation(s)
- Maryam Hajiabbas
- Laboratory of Pathophysiological and Nutritional Biochemistry, Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Blg G/E CP 611, B-1070, Brussels, Belgium
| | - Claudia D'Agostino
- Laboratory of Pathophysiological and Nutritional Biochemistry, Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Blg G/E CP 611, B-1070, Brussels, Belgium
| | - Julia Simińska-Stanny
- Department of Process Engineering and Technology of Polymer and Carbon Materials, Faculty of Chemistry, Wroclaw University of Science and Technology, Norwida 4/6, 50-373, Wroclaw, Poland.,3BIO-BioMatter, École Polytechnique de Bruxelles, Université Libre de Bruxelles, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
| | - Simon D Tran
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, H3A 0C7, Canada
| | - Amin Shavandi
- 3BIO-BioMatter, École Polytechnique de Bruxelles, Université Libre de Bruxelles, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
| | - Christine Delporte
- Laboratory of Pathophysiological and Nutritional Biochemistry, Faculty of Medicine, Université Libre de Bruxelles, 808 Route de Lennik, Blg G/E CP 611, B-1070, Brussels, Belgium.
| |
Collapse
|
28
|
Hayashi T, Eto K, Kadoya Y. Downregulation of ten-eleven translocation-2 triggers epithelial differentiation during organogenesis. Differentiation 2022; 125:45-53. [DOI: 10.1016/j.diff.2022.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/25/2022] [Accepted: 05/02/2022] [Indexed: 11/26/2022]
|
29
|
Shindo Y, Nakamura HM, Nakai J, Wakamori M, Nakamura T. Induction of myoepithelial cell differentiation by carbachol, a parasympathetic neurotransmitter agonist, during salivary gland development. Exp Cell Res 2022; 416:113137. [DOI: 10.1016/j.yexcr.2022.113137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/28/2022] [Accepted: 04/02/2022] [Indexed: 12/01/2022]
|
30
|
Moskwa N, Mahmood A, Nelson DA, Altrieth AL, Forni PE, Larsen M. Single-cell RNA sequencing reveals PDGFRα+ stromal cell subpopulations that promote proacinar cell differentiation in embryonic salivary gland organoids. Development 2022; 149:dev200167. [PMID: 35224622 PMCID: PMC8977102 DOI: 10.1242/dev.200167] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 02/18/2022] [Indexed: 12/12/2022]
Abstract
Stromal cells can direct the differentiation of epithelial progenitor cells during organ development. Fibroblast growth factor (FGF) signaling is essential for submandibular salivary gland development. Through stromal fibroblast cells, FGF2 can indirectly regulate proacinar cell differentiation in organoids, but the mechanisms are not understood. We performed single-cell RNA-sequencing and identified multiple stromal cell subsets, including Pdgfra+ stromal subsets expressing both Fgf2 and Fgf10. When combined with epithelial progenitor cells in organoids, magnetic-activated cell-sorted PDGFRα+ cells promoted proacinar cell differentiation similarly to total stroma. Gene expression analysis revealed that FGF2 increased the expression of multiple stromal genes, including Bmp2 and Bmp7. Both BMP2 and BMP7 synergized with FGF2, stimulating proacinar cell differentiation but not branching. However, stromal cells grown without FGF2 did not support proacinar organoid differentiation and instead differentiated into myofibroblasts. In organoids, TGFβ1 treatment stimulated myofibroblast differentiation and inhibited the proacinar cell differentiation of epithelial progenitor cells. Conversely, FGF2 reversed the effects of TGFβ1. We also demonstrated that adult salivary stromal cells were FGF2 responsive and could promote proacinar cell differentiation. These FGF2 signaling pathways may have applications in future regenerative therapies.
Collapse
Affiliation(s)
- Nicholas Moskwa
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA
- Graduate Program in Molecular, Cellular, Developmental and Neural Biology, University at Albany, State University of New York, Albany, NY 12222, USA
| | - Ayma Mahmood
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA
| | - Deirdre A. Nelson
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA
| | - Amber L. Altrieth
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA
- Graduate Program in Molecular, Cellular, Developmental and Neural Biology, University at Albany, State University of New York, Albany, NY 12222, USA
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
| | - Paolo E. Forni
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA
- Graduate Program in Molecular, Cellular, Developmental and Neural Biology, University at Albany, State University of New York, Albany, NY 12222, USA
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
| | - Melinda Larsen
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA
- Graduate Program in Molecular, Cellular, Developmental and Neural Biology, University at Albany, State University of New York, Albany, NY 12222, USA
- The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
| |
Collapse
|
31
|
Kim J, Eom MR, Ji Jeong E, Choi JS, Kwon SK. Multiple stimulation with spheroids comprising salivary gland and adipose-derived stem cells enhances regeneration of radiation-damaged salivary glands. J IND ENG CHEM 2022. [DOI: 10.1016/j.jiec.2021.12.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
32
|
Clark JF, Soriano PM. Pulling back the curtain: The hidden functions of receptor tyrosine kinases in development. Curr Top Dev Biol 2022; 149:123-152. [PMID: 35606055 PMCID: PMC9127239 DOI: 10.1016/bs.ctdb.2021.12.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Receptor tyrosine kinases (RTKs) are a conserved superfamily of transmembrane growth factor receptors that drive numerous cellular processes during development and in the adult. Upon activation, multiple adaptors and signaling effector proteins are recruited to binding site motifs located within the intracellular domain of the RTK. These RTK-effector interactions drive subsequent intracellular signaling cascades involved in canonical RTK signaling. Genetic dissection has revealed that alleles of Fibroblast Growth Factor receptors (FGFRs) that lack all canonical RTK signaling still retain some kinase-dependent biological activity. Here we examine how genetic analysis can be used to understand the mechanism by which RTKs drive multiple developmental processes via canonical signaling while revealing noncanonical activities. Recent data from both FGFRs and other RTKs highlight potential noncanonical roles in cell adhesion and nuclear signaling. The data supporting such functions are discussed as are recent technologies that have the potential to provide valuable insight into the developmental significance of these noncanonical activities.
Collapse
Affiliation(s)
- James F Clark
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Philippe M Soriano
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
33
|
Hou S, Zhu J, Shibata S, Nakamoto A, Kumano G. Repetitive accumulation of interstitial cells generates the branched structure of Cladonema medusa tentacles. Development 2021; 148:272708. [PMID: 34738619 DOI: 10.1242/dev.199544] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 10/26/2021] [Indexed: 12/20/2022]
Abstract
The shaping of tissues and organs in many animals relies on interactions between the epithelial cell layer and its underlying mesoderm-derived tissues. Inductive signals, such as receptor tyrosine kinase (RTK) signaling emanating from mesoderm, act on cells of the epithelium to initiate three-dimensional changes. However, how tissues are shaped in a diploblastic animal with no mesoderm remains largely unknown. In this study, the jellyfish Cladonema pacificum was used to investigate branch formation. The tentacles on its medusa stage undergo branching, which increases the epithelial surface area available for carrying nematocytes, thereby maximizing prey capture. Pharmacological and cellular analyses of the branching process suggest a two-step model for tentacle branch formation, in which mitogen-activated protein kinase kinase signaling accumulates interstitial cells in the future branch-forming region, and fibroblast growth factor signaling regulates branch elongation. This study highlights an essential role for these pluripotent stem cells in the tissue-shaping morphogenesis of a diploblastic animal. In addition, it identifies a mechanism involving RTK signaling and cell proliferative activity at the branch tip for branching morphogenesis that is apparently conserved across the animal kingdom.
Collapse
Affiliation(s)
- Shiting Hou
- Asamushi Research Center for Marine Biology, Graduate School of Life Sciences, Tohoku University, 9 Sakamoto, Asamushi, Aomori 039-3501, Japan
| | - Jianrong Zhu
- Asamushi Research Center for Marine Biology, Graduate School of Life Sciences, Tohoku University, 9 Sakamoto, Asamushi, Aomori 039-3501, Japan
| | - Saki Shibata
- Asamushi Research Center for Marine Biology, Graduate School of Life Sciences, Tohoku University, 9 Sakamoto, Asamushi, Aomori 039-3501, Japan
| | - Ayaki Nakamoto
- Asamushi Research Center for Marine Biology, Graduate School of Life Sciences, Tohoku University, 9 Sakamoto, Asamushi, Aomori 039-3501, Japan
| | - Gaku Kumano
- Asamushi Research Center for Marine Biology, Graduate School of Life Sciences, Tohoku University, 9 Sakamoto, Asamushi, Aomori 039-3501, Japan
| |
Collapse
|
34
|
Patel VN, Pineda DL, Berenstein E, Hauser BR, Choi S, Prochazkova M, Zheng C, Goldsmith CM, van Kuppevelt TH, Kulkarni A, Song Y, Linhardt RJ, Chibly AM, Hoffman MP. Loss of Hs3st3a1 or Hs3st3b1 enzymes alters heparan sulfate to reduce epithelial morphogenesis and adult salivary gland function. Matrix Biol 2021; 103-104:37-57. [PMID: 34653670 DOI: 10.1016/j.matbio.2021.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 09/10/2021] [Accepted: 10/04/2021] [Indexed: 12/25/2022]
Abstract
Heparan sulfate 3-O-sulfotransferases generate highly sulfated but rare 3-O-sulfated heparan sulfate (HS) epitopes on cell surfaces and in the extracellular matrix. Previous ex vivo experiments suggested functional redundancy exists among the family of seven enzymes but that Hs3st3a1 and Hs3st3b1 sulfated HS increases epithelial FGFR signaling and morphogenesis. Single-cell RNAseq analysis of control SMGs identifies increased expression of Hs3st3a1 and Hs3st3b1 in endbud and myoepithelial cells, both of which are progenitor cells during development and regeneration. To analyze their in vivo functions, we generated both Hs3st3a1-/- and Hs3st3b1-/- single knockout mice, which are viable and fertile. Salivary glands from both mice have impaired fetal epithelial morphogenesis when cultured with FGF10. Hs3st3b1-/- mice have reduced intact SMG branching morphogenesis and reduced 3-O-sulfated HS in the basement membrane. Analysis of HS biosynthetic enzyme transcription highlighted some compensatory changes in sulfotransferases expression early in development. The overall glycosaminoglycan composition of adult control and KO mice were similar, although HS disaccharide analysis showed increased N- and non-sulfated disaccharides in Hs3st3a1-/- HS. Analysis of adult KO gland function revealed normal secretory innervation, but without stimulation there was an increase in frequency of drinking behavior in both KO mice, suggesting basal salivary hypofunction, possibly due to myoepithelial dysfunction. Understanding how 3-O-sulfation regulates myoepithelial progenitor function will be important to manipulate HS-binding growth factors to enhance tissue function and regeneration.
Collapse
Affiliation(s)
- Vaishali N Patel
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dallas L Pineda
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elsa Berenstein
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Belinda R Hauser
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sophie Choi
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michaela Prochazkova
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Changyu Zheng
- Translational Research Core, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Corinne M Goldsmith
- Translational Research Core, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Toin H van Kuppevelt
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud university medical Centre, Nijmegen, Netherlands
| | - Ashok Kulkarni
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yuefan Song
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA
| | - Robert J Linhardt
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA
| | - Alejandro M Chibly
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Matthew P Hoffman
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
35
|
Clark JF, Ciccarelli EJ, Kayastha P, Ranepura G, Yamamoto KK, Hasan MS, Madaan U, Meléndez A, Savage-Dunn C. BMP pathway regulation of insulin signaling components promotes lipid storage in Caenorhabditis elegans. PLoS Genet 2021; 17:e1009836. [PMID: 34634043 PMCID: PMC8530300 DOI: 10.1371/journal.pgen.1009836] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/21/2021] [Accepted: 09/27/2021] [Indexed: 11/18/2022] Open
Abstract
A small number of peptide growth factor ligands are used repeatedly in development and homeostasis to drive programs of cell differentiation and function. Cells and tissues must integrate inputs from these diverse signals correctly, while failure to do so leads to pathology, reduced fitness, or death. Previous work using the nematode C. elegans identified an interaction between the bone morphogenetic protein (BMP) and insulin/IGF-1-like signaling (IIS) pathways in the regulation of lipid homeostasis. The molecular components required for this interaction, however, were not fully understood. Here we report that INS-4, one of 40 insulin-like peptides (ILPs), is regulated by BMP signaling to modulate fat accumulation. Furthermore, we find that the IIS transcription factor DAF-16/FoxO, but not SKN-1/Nrf, acts downstream of BMP signaling in lipid homeostasis. Interestingly, BMP activity alters sensitivity of these two transcription factors to IIS-promoted cytoplasmic retention in opposite ways. Finally, we probe the extent of BMP and IIS interactions by testing additional IIS functions including dauer formation, aging, and autophagy induction. Coupled with our previous work and that of other groups, we conclude that BMP and IIS pathways have at least three modes of interaction: independent, epistatic, and antagonistic. The molecular interactions we identify provide new insight into mechanisms of signaling crosstalk and potential therapeutic targets for IIS-related pathologies such as diabetes and metabolic syndrome.
Collapse
Affiliation(s)
- James F. Clark
- Biology Department, Queens College, City University of New York (CUNY), New York City, New York, United States of America
- Ph.D. Program in Biology, The Graduate Center, City University of New York (CUNY), New York City, New York, United States of America
| | - Emma J. Ciccarelli
- Biology Department, Queens College, City University of New York (CUNY), New York City, New York, United States of America
- Ph.D. Program in Biology, The Graduate Center, City University of New York (CUNY), New York City, New York, United States of America
| | - Peter Kayastha
- Biology Department, Queens College, City University of New York (CUNY), New York City, New York, United States of America
| | - Gehan Ranepura
- Biology Department, Queens College, City University of New York (CUNY), New York City, New York, United States of America
| | - Katerina K. Yamamoto
- Biology Department, Queens College, City University of New York (CUNY), New York City, New York, United States of America
- Ph.D. Program in Biology, The Graduate Center, City University of New York (CUNY), New York City, New York, United States of America
| | - Muhammad S. Hasan
- Biology Department, Queens College, City University of New York (CUNY), New York City, New York, United States of America
| | - Uday Madaan
- Biology Department, Queens College, City University of New York (CUNY), New York City, New York, United States of America
- Ph.D. Program in Biology, The Graduate Center, City University of New York (CUNY), New York City, New York, United States of America
| | - Alicia Meléndez
- Biology Department, Queens College, City University of New York (CUNY), New York City, New York, United States of America
- Ph.D. Program in Biology, The Graduate Center, City University of New York (CUNY), New York City, New York, United States of America
| | - Cathy Savage-Dunn
- Biology Department, Queens College, City University of New York (CUNY), New York City, New York, United States of America
- Ph.D. Program in Biology, The Graduate Center, City University of New York (CUNY), New York City, New York, United States of America
- * E-mail:
| |
Collapse
|
36
|
Kim D, Yoon YJ, Choi D, Kim J, Lim JY. 3D Organoid Culture From Adult Salivary Gland Tissues as an ex vivo Modeling of Salivary Gland Morphogenesis. Front Cell Dev Biol 2021; 9:698292. [PMID: 34458260 PMCID: PMC8397473 DOI: 10.3389/fcell.2021.698292] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022] Open
Abstract
Lumen formation of salivary glands has been investigated using in vivo or ex vivo rudiment culture models. In this study, we used a three-dimensional (3D) salivary gland organoid culture system and demonstrated that lumen formation could be recapitulated in mouse SMG organoids. In our organoid culture system, lumen formation was induced by vasoactive intestinal peptide and accelerated by treatment with RA. Furthermore, lumen formation was observed in branching duct-like structure when cultured in combination of fibroblast growth factors (FGF) in the presence of retinoic acid (RA). We suggest RA signaling-mediated regulation of VIPR1 and KRT7 as the underlying mechanism for lumen formation, rather than apoptosis in the organoid culture system. Collectively, our results support a fundamental role for RA in lumen formation and demonstrate the feasibility of 3D organoid culture as a tool for studying salivary gland morphogenesis.
Collapse
Affiliation(s)
- Donghyun Kim
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, South Korea
| | - Yeo-Jun Yoon
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, South Korea
| | - Dojin Choi
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, South Korea
| | - Jisun Kim
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, South Korea
| | - Jae-Yol Lim
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
37
|
Piraino LR, Benoit DSW, DeLouise LA. Salivary Gland Tissue Engineering Approaches: State of the Art and Future Directions. Cells 2021; 10:1723. [PMID: 34359893 PMCID: PMC8303463 DOI: 10.3390/cells10071723] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 01/08/2023] Open
Abstract
Salivary gland regeneration is important for developing treatments for radiation-induced xerostomia, Sjögren's syndrome, and other conditions that cause dry mouth. Culture conditions adopted from tissue engineering strategies have been used to recapitulate gland structure and function to study and regenerate the salivary glands. The purpose of this review is to highlight current trends in the field, with an emphasis on soluble factors that have been shown to improve secretory function in vitro. A PubMed search was conducted to identify articles published in the last 10 years and articles were evaluated to identify the most promising approaches and areas for further research. Results showed increasing use of extracellular matrix mimetics, such as Matrigel®, collagen, and a variety of functionalized polymers. Soluble factors that provide supportive cues, including fibroblast growth factors (FGFs) and neurotrophic factors, as well as chemical inhibitors of Rho-associated kinase (ROCK), epidermal growth factor receptor (EGFR), and transforming growth factor β receptor (TGFβR) have shown increases in important markers including aquaporin 5 (Aqp5); muscle, intestine, and stomach expression 1 (Mist1); and keratin (K5). However, recapitulation of tissue function at in vivo levels is still elusive. A focus on identification of soluble factors, cells, and/or matrix cues tested in combination may further increase the maintenance of salivary gland secretory function in vitro. These approaches may also be amenable for translation in vivo to support successful regeneration of dysfunctional glands.
Collapse
Affiliation(s)
- Lindsay R. Piraino
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, USA; (L.R.P.); (D.S.W.B.)
| | - Danielle S. W. Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, USA; (L.R.P.); (D.S.W.B.)
- Center for Oral Biology, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
- Materials Science Program, University of Rochester, Rochester, NY 14627, USA
- Department of Chemical Engineering, University of Rochester, Rochester, NY 14627, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Lisa A. DeLouise
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, USA; (L.R.P.); (D.S.W.B.)
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Materials Science Program, University of Rochester, Rochester, NY 14627, USA
- Department of Dermatology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
38
|
Wang S, Matsumoto K, Lish SR, Cartagena-Rivera AX, Yamada KM. Budding epithelial morphogenesis driven by cell-matrix versus cell-cell adhesion. Cell 2021; 184:3702-3716.e30. [PMID: 34133940 PMCID: PMC8287763 DOI: 10.1016/j.cell.2021.05.015] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 03/19/2021] [Accepted: 05/11/2021] [Indexed: 01/01/2023]
Abstract
Many embryonic organs undergo epithelial morphogenesis to form tree-like hierarchical structures. However, it remains unclear what drives the budding and branching of stratified epithelia, such as in the embryonic salivary gland and pancreas. Here, we performed live-organ imaging of mouse embryonic salivary glands at single-cell resolution to reveal that budding morphogenesis is driven by expansion and folding of a distinct epithelial surface cell sheet characterized by strong cell-matrix adhesions and weak cell-cell adhesions. Profiling of single-cell transcriptomes of this epithelium revealed spatial patterns of transcription underlying these cell adhesion differences. We then synthetically reconstituted budding morphogenesis by experimentally suppressing E-cadherin expression and inducing basement membrane formation in 3D spheroid cultures of engineered cells, which required β1-integrin-mediated cell-matrix adhesion for successful budding. Thus, stratified epithelial budding, the key first step of branching morphogenesis, is driven by an overall combination of strong cell-matrix adhesion and weak cell-cell adhesion by peripheral epithelial cells.
Collapse
Affiliation(s)
- Shaohe Wang
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA.
| | - Kazue Matsumoto
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Samantha R Lish
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Alexander X Cartagena-Rivera
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Kenneth M Yamada
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
39
|
Chatzeli L, Teshima THN, Hajihosseini MK, Gaete M, Proctor GB, Tucker AS. Comparing development and regeneration in the submandibular gland highlights distinct mechanisms. J Anat 2021; 238:1371-1385. [PMID: 33455001 PMCID: PMC8128775 DOI: 10.1111/joa.13387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/29/2022] Open
Abstract
A common question in organ regeneration is the extent to which regeneration recapitulates embryonic development. To investigate this concept, we compared the expression of two highly interlinked and essential genes for salivary gland development, Sox9 and Fgf10, during submandibular gland development, homeostasis and regeneration. Salivary gland duct ligation/deligation model was used as a regenerative model. Fgf10 and Sox9 expression changed during regeneration compared to homeostasis, suggesting that these key developmental genes play important roles during regeneration, however, significantly both displayed different patterns of expression in the regenerating gland compared to the developing gland. Regenerating glands, which during homeostasis had very few weakly expressing Sox9-positive cells in the striated/granular ducts, displayed elevated expression of Sox9 within these ducts. This pattern is in contrast to embryonic development, where Sox9 expression was absent in the proximally developing ducts. However, similar to the elevated expression at the distal tip of the epithelium in developing salivary glands, regenerating glands displayed elevated expression in a subpopulation of acinar cells, which during homeostasis expressed Sox9 at lower levels. A shift in expression of Fgf10 was observed from a widespread mesenchymal pattern during organogenesis to a more limited and predominantly epithelial pattern during homeostasis in the adult. This restricted expression in epithelial cells was maintained during regeneration, with no clear upregulation in the surrounding mesenchyme, as might be expected if regeneration recapitulated development. As both Fgf10 and Sox9 were upregulated in proximal ducts during regeneration, this suggests that the positive regulation of Sox9 by Fgf10, essential during development, is partially reawakened during regeneration using this model. Together these data suggest that developmentally important genes play a key role in salivary gland regeneration but do not precisely mimic the roles observed during development.
Collapse
Affiliation(s)
- Lemonia Chatzeli
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonUK
| | - Tathyane H. N. Teshima
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonUK
- Department of Oral MedicineUCL Eastman Dental InstituteLondonUK
| | | | - Marcia Gaete
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonUK
- Department of AnatomyFaculty of MedicinePontificia Universidad Católica de ChileSantiagoChile
| | - Gordon B. Proctor
- Centre for Host‐Microbiome InteractionsKing's College of LondonLondonUK
| | - Abigail S. Tucker
- Centre for Craniofacial and Regenerative BiologyKing's College LondonLondonUK
| |
Collapse
|
40
|
Efficient Surface Immobilization of Chemically Modified Hyaluronans for Enhanced Bioactivity and Survival of In Vitro-Cultured Embryonic Salivary Gland Mesenchymal Cells. Polymers (Basel) 2021; 13:polym13081216. [PMID: 33918722 PMCID: PMC8069509 DOI: 10.3390/polym13081216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/03/2021] [Accepted: 04/06/2021] [Indexed: 11/17/2022] Open
Abstract
Embryonic salivary gland mesenchyme (eSGM) secretes various growth factors (bioactives) that support the proper growth and differentiation of salivary gland epithelium. Therefore, eSGM cells can be used as feeder cells for in vitro-cultured artificial salivary gland if their survival and bioactivity are properly maintained. As eSGM is encapsulated in a hyaluronan (HA)-rich developmental milieu, we hypothesized that mimicking this environment in vitro via surface immobilization of HA might enhance survival and bioactivity of eSGM. In this study, various HA derivatives, conjugated with catechol (HA–CA), thiol (HA–SH), or amine (HA–EDA) moieties, respectively, were screened for their efficacy of culturing eSGM-derived feeder cells in vitro. Among these HA derivatives, HA–CA showed the highest surface coating efficiency and growth enhancement effect on the embryonic submandibular gland. In addition, the HA–CA coating enhanced the production of growth factors EGF and FGF7, but not FGF10. These effects were maintained when eSGM cells isolated from the embryonic salivary gland were re-seeded to develop the feeder layer cells. CD44s (a major HA receptor) in eSGM cells were clustered at the cell membrane, and enhanced EGF expression was detected only in CD44 cluster-positive cells, suggesting that membrane clustering of CD44 is the key mechanism for the increased expression of EGF.
Collapse
|
41
|
Watanabe T, Kometani-Gunjigake K, Nakao-Kuroishi K, Ito-Sago M, Mizuhara M, Iwata D, Moriyama K, Ono K, Kawamoto T. A Ser252Trp substitution in mouse FGFR2 results in hyperplasia of embryonic salivary gland parenchyma. J Oral Biosci 2021; 63:184-191. [PMID: 33684522 DOI: 10.1016/j.job.2021.02.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/24/2021] [Accepted: 01/30/2021] [Indexed: 10/22/2022]
Abstract
OBJECTIVES Mutations in the fibroblast growth factor receptor 2 (FGFR2) gene are responsible for several severe forms of craniosynostotic disorders, such as Apert and Crouzon syndromes. Patients with craniosynostotic disorders caused by a mutation in Fgfr2 present with several clinical symptoms, including hypersalivation. Here we used a transgenic mouse model of Apert syndrome (Fgfr2+/S252W mice) to evaluate the morphology of the submandibular glands at embryonic day 15.5 (E15.5), the time point reported to mark the start of lumen formation. METHODS Fgfr2+/S252W mice were generated by crossing ACTB-Cre+/+ and Fgfr2+/Neo-S252W mice. After measuring body weight, the submandibular glands were collected at E15.5. H&E staining, immunostaining, and RT-qPCR were performed to investigate the development of the submandibular gland. RESULTS The number of ducts and acini in Fgfr2+/S252W mice was significantly higher than in control littermates; however, lumen formation was not affected. The mRNA expression of Fgf1, Fgfr1, Mmp2, Bmp4, Bmp7, Dusp6, and Etv5 in Fgfr2+/S252W mice was significantly higher compared to control littermates. Immunoreactivity for FGF3, FGF1, BMP4, and F4/80 was detected in the parenchyma of Fgfr2+/S252W mice. The area of apoptotic cells stained with TUNEL in Fgfr2+/S252W mice was significantly larger than that of the control littermates. CONCLUSIONS These results suggested that increased FGFR1 signaling and apoptosis in the submandibular glands of Fgfr2+/S252W mice occurred at E15.5, leading to parenchymal hyperplasia. This study demonstrated that a Ser252Trp substitution in mouse FGFR2 resulted in hyperplasia of the submandibular gland parenchyma during development.
Collapse
Affiliation(s)
- Tsukasa Watanabe
- Division of Orofacial Functions and Orthodontics, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka, 803-8580, Japan
| | - Kaori Kometani-Gunjigake
- Division of Orofacial Functions and Orthodontics, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka, 803-8580, Japan
| | - Kayoko Nakao-Kuroishi
- Division of Orofacial Functions and Orthodontics, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka, 803-8580, Japan
| | - Misa Ito-Sago
- Division of Orofacial Functions and Orthodontics, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka, 803-8580, Japan
| | - Masahiro Mizuhara
- Division of Orofacial Functions and Orthodontics, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka, 803-8580, Japan
| | - Daiki Iwata
- Division of Orofacial Functions and Orthodontics, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka, 803-8580, Japan
| | - Keiji Moriyama
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Kentaro Ono
- Division of Physiology, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka, 803-8580, Japan
| | - Tatsuo Kawamoto
- Division of Orofacial Functions and Orthodontics, Kyushu Dental University, 2-6-1 Manazuru, Kokurakita-ku, Kitakyushu, Fukuoka, 803-8580, Japan.
| |
Collapse
|
42
|
Barbosa GO, Biancardi MF, Carvalho HF. Heparan sulfate fine‐tunes stromal‐epithelial communication in the prostate gland. Dev Dyn 2020; 250:618-628. [DOI: 10.1002/dvdy.281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/20/2020] [Accepted: 12/10/2020] [Indexed: 12/19/2022] Open
Affiliation(s)
- Guilherme O. Barbosa
- Department of Structural and Functional Biology, Institute of Biology State University of Campinas Campinas Brazil
| | - Manoel F. Biancardi
- Department of Histology, Embryology and Cell Biology, Institute of Biological Sciences Federal University of Goiás Goiânia Brazil
| | - Hernandes F. Carvalho
- Department of Structural and Functional Biology, Institute of Biology State University of Campinas Campinas Brazil
| |
Collapse
|
43
|
Li M, Qi L, Xu JB, Zhong LY, Chan S, Chen SN, Shao XR, Zheng LY, Dong ZX, Fang TL, Mai ZY, Li J, Zheng Y, Zhang XD. Methylation of the Promoter Region of the Tight Junction Protein-1 by DNMT1 Induces EMT-like Features in Multiple Myeloma. MOLECULAR THERAPY-ONCOLYTICS 2020; 19:197-207. [PMID: 33251332 PMCID: PMC7666313 DOI: 10.1016/j.omto.2020.10.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/07/2020] [Indexed: 11/19/2022]
Abstract
The molecular alterations that initiate the development of multiple myeloma (MM) are not fully understood. Our results revealed that TJP1 was downregulated in MM and positively related to the overall survival of MM patients in The Cancer Genome Atlas (TCGA) database and patient samples. In parallel, cell adhesion capacity representing MM metastasis was decreased in MM patients compared with healthy samples, together with the significantly activated epithelial-to-mesenchymal transition (EMT) transcriptional-like patterns of MM cells. Further analyses demonstrated that TJP1 negatively regulated EMT and consequently positively regulated cell adhesion in MM from TCGA database and MM1s cells. Furthermore, the methylation level of each CpG site on the TJP1 promoter was negatively correlated with TJP1 expression levels. Quantitative real-time PCR and western blot assays demonstrated that methylase DNMT1 regulated the methylation of TJP1. Finally, treatment with a combination of the MM clinical medicine bortezomib, methylation inhibitor, or TJP1 overexpression significantly suppressed the viability and progression of tumor cells of MM orthotopic models. In summary, our results indicate that DNMT1 promotes the methylation of TJP1 promoter, thereby decreasing its expression and regulating the development of EMT-inhibited MM cell adhesion. Therefore, methylation of TJP1 is a potential therapeutic agent to prevent the progression of MM disease.
Collapse
Affiliation(s)
- Miao Li
- Department of Hematology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
- Department of Pharmacology, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen 518107, China
| | - Lin Qi
- Department of Pharmacology, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen 518107, China
- Corresponding author: Lin Qi, Department of Pharmacology, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen 518107, China.
| | - Jing-Bo Xu
- Department of Hematology, Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| | - Li-Ye Zhong
- Department of Hematology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510000, China
| | - Szehoi Chan
- Department of Pharmacology, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen 518107, China
| | - Shu-Na Chen
- Department of Pharmacology, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen 518107, China
| | - Xin-Rong Shao
- Department of Pharmacology, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen 518107, China
| | - Li-Yuan Zheng
- Department of Pharmacology, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen 518107, China
| | - Zhao-Xia Dong
- Department of Pharmacology, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen 518107, China
| | - Tian-Liang Fang
- Department of Pharmacology, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen 518107, China
| | - Zhi-Ying Mai
- Department of Hematology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
- Department of Pharmacology, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen 518107, China
| | - Juan Li
- Department of Hematology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
| | - Yongjiang Zheng
- Department of Hematology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
- Corresponding author: Yongjiang Zheng, Department of Hematology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Xing-Ding Zhang
- Department of Pharmacology, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen 518107, China
- Corresponding author: Xing-Ding Zhang, Department of Pharmacology, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen 518107, China.
| |
Collapse
|
44
|
Barrows CM, Wu D, Farach-Carson MC, Young S. Building a Functional Salivary Gland for Cell-Based Therapy: More than Secretory Epithelial Acini. Tissue Eng Part A 2020; 26:1332-1348. [PMID: 32829674 PMCID: PMC7759264 DOI: 10.1089/ten.tea.2020.0184] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/20/2020] [Indexed: 11/13/2022] Open
Abstract
A few treatment options exist for patients experiencing xerostomia due to hyposalivation that occurs as a result of disease or injury to the gland. An opportunity for a permanent solution lies in the field of salivary gland replacement through tissue engineering. Recent success emboldens in the vision of producing a tissue-engineered salivary gland composed of differentiated salivary epithelial cells that are able to differentiate to form functional units that produce and deliver saliva to the oral cavity. This vision is augmented by advances in understanding cellular mechanisms that guide branching morphogenesis and salivary epithelial cell polarization in both acinar and ductal structures. Growth factors and other guidance cues introduced into engineered constructs help to develop a more complex glandular structure that seeks to mimic native salivary gland tissue. This review describes the separate epithelial phenotypes that make up the gland, and it describes their relationship with the other cell types such as nerve and vasculature that surround them. The review is organized around the links between the native components that form and contribute to various aspects of salivary gland development, structure, and function and how this information can drive the design of functional tissue-engineered constructs. In addition, we discuss the attributes of various biomaterials commonly used to drive function and form in engineered constructs. The review also contains a current description of the state-of-the-art of the field, including successes and challenges in creating materials for preclinical testing in animal models. The ability to integrate biomolecular cues in combination with a range of materials opens the door to the design of increasingly complex salivary gland structures that, once accomplished, can lead to breakthroughs in other fields of tissue engineering of epithelial-based exocrine glands or oral tissues.
Collapse
Affiliation(s)
- Caitlynn M.L. Barrows
- Department of Diagnostic and Biomedical Sciences and The University of Texas Health Science Center at Houston, School of Dentistry, Houston, Texas, USA
- Department of Oral and Maxillofacial Surgery, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, Texas, USA
| | - Danielle Wu
- Department of Diagnostic and Biomedical Sciences and The University of Texas Health Science Center at Houston, School of Dentistry, Houston, Texas, USA
| | - Mary C. Farach-Carson
- Department of Diagnostic and Biomedical Sciences and The University of Texas Health Science Center at Houston, School of Dentistry, Houston, Texas, USA
- Department of Biosciences and Rice University, Houston, Texas, USA
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Simon Young
- Department of Oral and Maxillofacial Surgery, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, Texas, USA
| |
Collapse
|
45
|
Ray AT, Mazot P, Brewer JR, Catela C, Dinsmore CJ, Soriano P. FGF signaling regulates development by processes beyond canonical pathways. Genes Dev 2020; 34:1735-1752. [PMID: 33184218 PMCID: PMC7706708 DOI: 10.1101/gad.342956.120] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 10/13/2020] [Indexed: 01/06/2023]
Abstract
FGFs are key developmental regulators that engage a signal transduction cascade through receptor tyrosine kinases, prominently engaging ERK1/2 but also other pathways. However, it remains unknown whether all FGF activities depend on this canonical signal transduction cascade. To address this question, we generated allelic series of knock-in Fgfr1 and Fgfr2 mouse strains, carrying point mutations that disrupt binding of signaling effectors, and a kinase dead allele of Fgfr2 that broadly phenocopies the null mutant. When interrogated in cranial neural crest cells, we identified discrete functions for signaling pathways in specific craniofacial contexts, but point mutations, even when combined, failed to recapitulate the single or double null mutant phenotypes. Furthermore, the signaling mutations abrogated established FGF-induced signal transduction pathways, yet FGF functions such as cell-matrix and cell-cell adhesion remained unaffected, though these activities did require FGFR kinase activity. Our studies establish combinatorial roles of Fgfr1 and Fgfr2 in development and uncouple novel FGFR kinase-dependent cell adhesion properties from canonical intracellular signaling.
Collapse
MESH Headings
- Animals
- Cell Adhesion/genetics
- Cell Death/genetics
- Cells, Cultured
- Fibroblast Growth Factors/physiology
- Gene Expression Regulation, Developmental/genetics
- Mice
- Mutation
- Neural Crest/cytology
- Protein Kinases/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
- Signal Transduction/genetics
Collapse
Affiliation(s)
- Ayan T Ray
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Pierre Mazot
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - J Richard Brewer
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Catarina Catela
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Colin J Dinsmore
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Philippe Soriano
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| |
Collapse
|
46
|
Xie Y, Su N, Yang J, Tan Q, Huang S, Jin M, Ni Z, Zhang B, Zhang D, Luo F, Chen H, Sun X, Feng JQ, Qi H, Chen L. FGF/FGFR signaling in health and disease. Signal Transduct Target Ther 2020; 5:181. [PMID: 32879300 PMCID: PMC7468161 DOI: 10.1038/s41392-020-00222-7] [Citation(s) in RCA: 470] [Impact Index Per Article: 94.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/28/2020] [Accepted: 06/15/2020] [Indexed: 12/13/2022] Open
Abstract
Growing evidences suggest that the fibroblast growth factor/FGF receptor (FGF/FGFR) signaling has crucial roles in a multitude of processes during embryonic development and adult homeostasis by regulating cellular lineage commitment, differentiation, proliferation, and apoptosis of various types of cells. In this review, we provide a comprehensive overview of the current understanding of FGF signaling and its roles in organ development, injury repair, and the pathophysiology of spectrum of diseases, which is a consequence of FGF signaling dysregulation, including cancers and chronic kidney disease (CKD). In this context, the agonists and antagonists for FGF-FGFRs might have therapeutic benefits in multiple systems.
Collapse
Affiliation(s)
- Yangli Xie
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| | - Nan Su
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Jing Yang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Qiaoyan Tan
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Shuo Huang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Min Jin
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhenhong Ni
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Bin Zhang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Dali Zhang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Fengtao Luo
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Hangang Chen
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Xianding Sun
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Jian Q Feng
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, 75246, USA
| | - Huabing Qi
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| | - Lin Chen
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
47
|
Ikai K, Sakai M, Minagi HO, Gojo N, Sakai T. ΔNp63 is upregulated during salivary gland regeneration following duct ligation and irradiation in mice. FEBS Lett 2020; 594:3216-3226. [PMID: 32748407 DOI: 10.1002/1873-3468.13896] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/30/2020] [Accepted: 07/21/2020] [Indexed: 01/12/2023]
Abstract
The transcription factor p63, a component of the p53 family, has important functions in development, homeostasis, and regeneration of epithelial tissues. However, the role of p63 in the regeneration of exocrine glands, including the salivary glands (SGs), has not been fully investigated. We investigated p63 expression in SG regeneration induced by duct ligation and irradiation. The expression of ΔNp63, a p63 isoform, increased and was colocalized with keratin 5 positive cells were myoepithelial cells. Furthermore, ΔNp63 expression was regulated by FGF7 stimulation via p38 MAPK phosphorylation and affected SG morphogenesis. These results suggest that ΔNp63 is essential for SG regeneration and may be a new target for regenerative treatment.
Collapse
Affiliation(s)
- Kazuki Ikai
- Department of Oral-facial Disorders, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Manabu Sakai
- Department of Clinical Laboratory, Osaka University Dental Hospital, Suita, Japan
| | - Hitomi Ono Minagi
- Department of Cytology and Histology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Nao Gojo
- Department of Oral-facial Disorders, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Takayoshi Sakai
- Department of Oral-facial Disorders, Osaka University Graduate School of Dentistry, Suita, Japan
| |
Collapse
|
48
|
Padmanaban V, Grasset EM, Neumann NM, Fraser AK, Henriet E, Matsui W, Tran PT, Cheung KJ, Georgess D, Ewald AJ. Organotypic culture assays for murine and human primary and metastatic-site tumors. Nat Protoc 2020; 15:2413-2442. [PMID: 32690957 PMCID: PMC8202162 DOI: 10.1038/s41596-020-0335-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 04/16/2020] [Indexed: 01/20/2023]
Abstract
Cancer invasion and metastasis are challenging to study in vivo since they occur deep inside the body over extended time periods. Organotypic 3D culture of fresh tumor tissue enables convenient real-time imaging, genetic and microenvironmental manipulation and molecular analysis. Here, we provide detailed protocols to isolate and culture heterogenous organoids from murine and human primary and metastatic site tumors. The time required to isolate organoids can vary based on the tissue and organ type but typically takes <7 h. We describe a suite of assays that model specific aspects of metastasis, including proliferation, survival, invasion, dissemination and colony formation. We also specify comprehensive protocols for downstream applications of organotypic cultures that will allow users to (i) test the role of specific genes in regulating various cellular processes, (ii) distinguish the contributions of several microenvironmental factors and (iii) test the effects of novel therapeutics.
Collapse
Affiliation(s)
- Veena Padmanaban
- Department of Cell Biology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Eloise M. Grasset
- Department of Cell Biology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Neil M. Neumann
- Department of Cell Biology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Andrew K. Fraser
- Department of Cell Biology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Elodie Henriet
- Department of Cell Biology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - William Matsui
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Phuoc T. Tran
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kevin J. Cheung
- Department of Cell Biology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA,Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Dan Georgess
- Department of Cell Biology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA,Department of Natural Sciences, School of Arts & Sciences, Lebanese American University, Beirut, Lebanon
| | - Andrew J. Ewald
- Department of Cell Biology, Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA,Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Author for Correspondence: Andrew J. Ewald, 855 N. Wolfe Street, Rangos 452, Baltimore, MD 21205, Tel: 410-614-9288,
| |
Collapse
|
49
|
Abstract
ABSTRACT
Over the past 5 years, several studies have begun to uncover the links between the classical signal transduction pathways and the physical mechanisms that are used to sculpt branched tissues. These advances have been made, in part, thanks to innovations in live imaging and reporter animals. With modern research tools, our conceptual models of branching morphogenesis are rapidly evolving, and the differences in branching mechanisms between each organ are becoming increasingly apparent. Here, we highlight four branched epithelia that develop at different spatial scales, within different surrounding tissues and via divergent physical mechanisms. Each of these organs has evolved to employ unique branching strategies to achieve a specialized final architecture.
Collapse
Affiliation(s)
- Katharine Goodwin
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Celeste M. Nelson
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
50
|
Tanaka J, Mishima K. In vitro three-dimensional culture systems of salivary glands. Pathol Int 2020; 70:493-501. [PMID: 32367629 DOI: 10.1111/pin.12947] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/20/2020] [Accepted: 04/22/2020] [Indexed: 12/18/2022]
Abstract
Dry mouth can be caused by salivary gland hypofunction due to Sjögren's syndrome (SS) or radiation therapy for head and neck cancer, and it can also be a side effect of medications. The use of sialagogues effectively increases saliva secretion in patients with dry mouth. However, the application of sialagogues is not always satisfactory because of their side effects, such as sweating, nausea, runny nose and diarrhea. Two-dimensional (2D) cell cultures have been used not only for drug screening and discovery but also to clarify disease mechanisms. However, three-dimensional (3D) cell cultures are expected to be even more advantageous than 2D cell cultures. Therefore, we have tried to develop an in vitro cell culture system that can reconstitute 3D salivary glands. Sox9 and Foxc1 were identified as important genes that differentiate mouse embryonic stem cell-derived oral ectoderm into salivary gland placode. Using these genes and organoid culture systems, we succeeded in generating salivary gland organoids that exhibited a morphology and gene expression profile that were similar to those of the embryonic rudiment from which salivary glands arise in normal mice. These organoids are expected to be a promising tool for disease modeling, drug discovery and regenerative medicine in salivary glands.
Collapse
Affiliation(s)
- Junichi Tanaka
- Division of Pathology, Department of Oral Diagnostic Sciences, Showa University School of Dentistry, Tokyo, Japan
| | - Kenji Mishima
- Division of Pathology, Department of Oral Diagnostic Sciences, Showa University School of Dentistry, Tokyo, Japan
| |
Collapse
|