1
|
Nauryzgaliyeva Z, Goux Corredera I, Garreta E, Montserrat N. Harnessing mechanobiology for kidney organoid research. Front Cell Dev Biol 2023; 11:1273923. [PMID: 38077999 PMCID: PMC10704179 DOI: 10.3389/fcell.2023.1273923] [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: 08/07/2023] [Accepted: 10/16/2023] [Indexed: 10/16/2024] Open
Abstract
Recently, organoids have emerged as revolutionizing tools with the unprecedented potential to recreate organ-specific microanatomy in vitro. Upon their derivation from human pluripotent stem cells (hPSCs), organoids reveal the blueprints of human organogenesis, further allowing the faithful recapitulation of their physiology. Nevertheless, along with the evolution of this field, advanced research exposed the organoids' shortcomings, particularly regarding poor reproducibility rates and overall immatureness. To resolve these challenges, many studies have started to underscore the relevance of mechanical cues as a relevant source to induce and externally control hPSCs differentiation. Indeed, established organoid generation protocols from hPSCs have mainly relyed on the biochemical induction of fundamental signalling pathways present during kidney formation in mammals, whereas mechanical cues have largely been unexplored. This review aims to discuss the pertinence of (bio) physical cues within hPSCs-derived organoid cultures, while deciphering their effect on morphogenesis. Moreover, we will explore state-of-the-art mechanobiology techniques as revolutionizing means for understanding the underlying role of mechanical forces in biological processes in organoid model systems.
Collapse
Affiliation(s)
- Zarina Nauryzgaliyeva
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Iphigénie Goux Corredera
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), University of Barcelona, Barcelona, Spain
| | - Elena Garreta
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), University of Barcelona, Barcelona, Spain
| | - Nuria Montserrat
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina, Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
2
|
Fowler EW, Witt RL, Jia X. Basement Membrane Mimetic Hydrogel Cooperates with Rho-Associated Protein Kinase Inhibitor to Promote the Development of Acini-Like Salivary Gland Spheroids. ADVANCED NANOBIOMED RESEARCH 2023; 3:2300088. [PMID: 38645834 PMCID: PMC11031203 DOI: 10.1002/anbr.202300088] [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] [Indexed: 04/23/2024] Open
Abstract
Successful engineering of functional salivary glands necessitates the creation of cell-instructive environments for ex vivo expansion and lineage specification of primary human salivary gland stem cells (hS/PCs). Herein, basement membrane mimetic hydrogels were prepared using hyaluronic acid, cell adhesive peptides, and hyperbranched polyglycerol (HPG), with or without sulfate groups, to produce "hyperGel+" or "hyperGel", respectively. Differential scanning fluorescence experiments confirmed the ability of the sulphated HPG precursor to stabilize fibroblast growth factor 10. The hydrogels were nanoporous, cytocompatibile and cell-permissive, enabling the development of multicellular hS/PC spheroids in 14 days. Incorporation of sulfated HPG species in the hydrogel enhanced cell proliferation. Culture of hS/PCs in hyperGel+ in the presence of a Rho kinase inhibitor, Y-27632 (Y-27), led to the development of spheroids with a central lumen, increased the expression of acinar marker aquaporin-3 at the transcript level (AQP3), and decreased the expression of ductal marker keratin 7 at both the transcript (KRT7) and the protein levels (K7). Reduced expression of transforming growth factor beta (TGF-β) targets SMAD2/3 was also observed in Y27-treated cultures, suggesting attenuation of TGF-β signaling. Thus, hyperGel+ cooperates with the ROCK inhibitor to promote the development of lumened spheroids with enhanced expression of acinar markers.
Collapse
Affiliation(s)
- Eric W. Fowler
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware, 19716, USA
| | - Robert L. Witt
- Helen F. Graham Cancer Center and Research Institute, Christiana Care, Newark, Delaware, 19713, USA
| | - Xinqiao Jia
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware, 19716, USA
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware, 19716, USA
- Delaware Biotechnology Institute, 590 Avenue 1743, Newark, DE 19713, USA
| |
Collapse
|
3
|
Chen G, Gao X, Chen J, Peng L, Chen S, Tang C, Dai Y, Wei Q, Luo D. Actomyosin Activity and Piezo1 Activity Synergistically Drive Urinary System Fibroblast Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303369. [PMID: 37867255 PMCID: PMC10667826 DOI: 10.1002/advs.202303369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 08/11/2023] [Indexed: 10/24/2023]
Abstract
Mechanical cues play a crucial role in activating myofibroblasts from quiescent fibroblasts during fibrosis, and the stiffness of the extracellular matrix is of significant importance in this process. While intracellular force mediated by myosin II and calcium influx regulated by Piezo1 are the primary mechanisms by which cells sense and respond to mechanical forces, their intercellular mechanical interaction remains to be elucidated. Here, hydrogels with tunable substrate are used to systematically investigate the crosstalk of myosin II and Piezo1 in fibroblast to myofibroblast transition (FMT). The findings reveal that the two distinct signaling pathways are integrated to convert mechanical stiffness signals into biochemical signals during bladder-specific FMT. Moreover, it is demonstrated that the crosstalk between myosin II and Piezo1 sensing mechanisms synergistically establishes a sustained feed-forward loop that contributes to chromatin remodeling, induces the expression of downstream target genes, and ultimately exacerbates FMT, in which the intracellular force activates Piezo1 by PI3K/PIP3 pathway-mediated membrane tension and the Piezo1-regulated calcium influx enhances intracellular force by the classical FAK/RhoA/ROCK pathway. Finally, the multifunctional Piezo1 in the complex feedback circuit of FMT drives to further identify that targeting Piezo1 as a therapeutic option for ameliorating bladder fibrosis and dysfunction.
Collapse
Affiliation(s)
- Guo Chen
- Department of UrologyInstitute of Urology (Laboratory of Reconstructive Urology)West China HospitalSichuan UniversityChengduSichuan610041P. R. China
- Department of Urology and Pelvic surgeryWest China School of Public Health and West China Fourth HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Xiaoshuai Gao
- Department of UrologyInstitute of Urology (Laboratory of Reconstructive Urology)West China HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Jiawei Chen
- Department of UrologyInstitute of Urology (Laboratory of Reconstructive Urology)West China HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Liao Peng
- Department of UrologyInstitute of Urology (Laboratory of Reconstructive Urology)West China HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Shuang Chen
- Department of UrologyInstitute of Urology (Laboratory of Reconstructive Urology)West China HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Cai Tang
- Department of UrologyInstitute of Urology (Laboratory of Reconstructive Urology)West China HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Yi Dai
- Department of Urology and Pelvic surgeryWest China School of Public Health and West China Fourth HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Qiang Wei
- College of Polymer Science and EngineeringState Key Laboratory of Polymer Materials and EngineeringSichuan UniversityChengduSichuan610065P. R. China
| | - Deyi Luo
- Department of UrologyInstitute of Urology (Laboratory of Reconstructive Urology)West China HospitalSichuan UniversityChengduSichuan610041P. R. China
| |
Collapse
|
4
|
Bonnet H, Isidro Alonso CA, Gupta IR. Submandibular gland epithelial development and the importance of junctions. Tissue Barriers 2023; 11:2161255. [PMID: 36576256 PMCID: PMC10606785 DOI: 10.1080/21688370.2022.2161255] [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: 07/03/2022] [Revised: 12/08/2022] [Accepted: 12/18/2022] [Indexed: 12/29/2022] Open
Abstract
Salivary glands consist of highly specialized epithelial cells that secrete the fluid, saliva, and/or transport saliva into the oral cavity. Saliva is essential to lubricate the oral cavity for food consumption and to maintain the hygiene of the oral cavity. In this review, we will focus on the formation of the epithelial cell lineage and the cell junctions that are essential for formation of saliva and maintenance of the epithelial barrier between the ducts that transport saliva and the extracellular environment.
Collapse
Affiliation(s)
| | - Carlos Agustin Isidro Alonso
- Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Indra R. Gupta
- Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
5
|
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: 4] [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: 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
|
6
|
Du W, Bhojwani A, Hu JK. FACEts of mechanical regulation in the morphogenesis of craniofacial structures. Int J Oral Sci 2021; 13:4. [PMID: 33547271 PMCID: PMC7865003 DOI: 10.1038/s41368-020-00110-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023] Open
Abstract
During embryonic development, organs undergo distinct and programmed morphological changes as they develop into their functional forms. While genetics and biochemical signals are well recognized regulators of morphogenesis, mechanical forces and the physical properties of tissues are now emerging as integral parts of this process as well. These physical factors drive coordinated cell movements and reorganizations, shape and size changes, proliferation and differentiation, as well as gene expression changes, and ultimately sculpt any developing structure by guiding correct cellular architectures and compositions. In this review we focus on several craniofacial structures, including the tooth, the mandible, the palate, and the cranium. We discuss the spatiotemporal regulation of different mechanical cues at both the cellular and tissue scales during craniofacial development and examine how tissue mechanics control various aspects of cell biology and signaling to shape a developing craniofacial organ.
Collapse
Affiliation(s)
- Wei Du
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Arshia Bhojwani
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Jimmy K Hu
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
7
|
Oyelakin A, Song E, Min S, Bard J, Kann J, Horeth E, Smalley K, Kramer J, Sinha S, Romano R. Transcriptomic and Single-Cell Analysis of the Murine Parotid Gland. J Dent Res 2019; 98:1539-1547. [PMID: 31623513 PMCID: PMC6873284 DOI: 10.1177/0022034519882355] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The salivary complex of mammals consists of 3 major pairs of glands: the parotid, submandibular, and sublingual glands. While the 3 glands share similar functional properties, such as saliva secretion, their differences are largely based on the types of secretions they produce. While recent studies have begun to shed light on the underlying molecular differences among the glands, few have examined the global transcriptional repertoire over various stages of gland maturation. To better elucidate the molecular nature of the parotid gland, we have performed RNA sequencing to generate comprehensive and global gene expression profiles of this gland at different stages of maturation. Our transcriptomic characterization and hierarchical clustering analysis with adult organ RNA sequencing data sets has identified a number of molecular players and pathways that are relevant for parotid gland biology. Moreover, our detailed analysis has revealed a unique parotid gland-specific gene signature that may represent important players that could impart parotid gland-specific biological properties. To complement our transcriptomic studies, we have performed single-cell RNA sequencing to map the transcriptomes of parotid epithelial cells. Interrogation of the single-cell transcriptomes revealed the degree of molecular and cellular heterogeneity of the various epithelial cell types within the parotid gland. Moreover, we uncovered a mixed-lineage population of cells that may reflect molecular priming of differentiation potentials. Overall our comprehensive studies provide a powerful tool for the discovery of novel molecular players important in parotid gland biology.
Collapse
Affiliation(s)
- A. Oyelakin
- Department of Oral Biology, School of Dental
Medicine, State University of New York at Buffalo, Buffalo, NY, USA
| | - E.A.C. Song
- Department of Oral Biology, School of Dental
Medicine, State University of New York at Buffalo, Buffalo, NY, USA
| | - S. Min
- Department of Oral Biology, School of Dental
Medicine, State University of New York at Buffalo, Buffalo, NY, USA
| | - J.E. Bard
- Genomics and Bioinformatics Core, State
University of New York at Buffalo, Buffalo, NY, USA
| | - J.V. Kann
- Genomics and Bioinformatics Core, State
University of New York at Buffalo, Buffalo, NY, USA
| | - E. Horeth
- Department of Oral Biology, School of Dental
Medicine, State University of New York at Buffalo, Buffalo, NY, USA
| | - K. Smalley
- Department of Biochemistry, Jacobs School of
Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY,
USA
| | - J.M. Kramer
- Department of Oral Biology, School of Dental
Medicine, State University of New York at Buffalo, Buffalo, NY, USA
| | - S. Sinha
- Department of Biochemistry, Jacobs School of
Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY,
USA
| | - R.A. Romano
- Department of Oral Biology, School of Dental
Medicine, State University of New York at Buffalo, Buffalo, NY, USA
- Department of Biochemistry, Jacobs School of
Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY,
USA
| |
Collapse
|
8
|
Donnelly H, Salmeron-Sanchez M, Dalby MJ. Designing stem cell niches for differentiation and self-renewal. J R Soc Interface 2019; 15:rsif.2018.0388. [PMID: 30158185 PMCID: PMC6127175 DOI: 10.1098/rsif.2018.0388] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Accepted: 08/08/2018] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells, characterized by their ability to differentiate into skeletal tissues and self-renew, hold great promise for both regenerative medicine and novel therapeutic discovery. However, their regenerative capacity is retained only when in contact with their specialized microenvironment, termed the stem cell niche Niches provide structural and functional cues that are both biochemical and biophysical, stem cells integrate this complex array of signals with intrinsic regulatory networks to meet physiological demands. Although, some of these regulatory mechanisms remain poorly understood or difficult to harness with traditional culture systems. Biomaterial strategies are being developed that aim to recapitulate stem cell niches, by engineering microenvironments with physiological-like niche properties that aim to elucidate stem cell-regulatory mechanisms, and to harness their regenerative capacity in vitro In the future, engineered niches will prove important tools for both regenerative medicine and therapeutic discoveries.
Collapse
Affiliation(s)
- Hannah Donnelly
- The Centre for the Cellular Microenvironment, University of Glasgow, Glasgow G12 8QQ, UK
| | | | - Matthew J Dalby
- The Centre for the Cellular Microenvironment, University of Glasgow, Glasgow G12 8QQ, UK
| |
Collapse
|
9
|
Wu D, Witt RL, Harrington DA, Farach-Carson MC. Dynamic Assembly of Human Salivary Stem/Progenitor Microstructures Requires Coordinated α 1β 1 Integrin-Mediated Motility. Front Cell Dev Biol 2019; 7:224. [PMID: 31750298 PMCID: PMC6843075 DOI: 10.3389/fcell.2019.00224] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 09/20/2019] [Indexed: 12/26/2022] Open
Abstract
A tissue engineering approach can provide replacement salivary gland structures to patients with hyposalivation disorders and xerostomia. Salivary human stem/progenitor cells (hS/PCs) were isolated from healthy regions of parotid glands of head and neck surgery patients, expanded, then encapsulated in biocompatible hyaluronate (HA)-based hydrogels. These bioactive hydrogels provide a surrogate territorial matrix suitable for the dynamic assembly, growth and reorganization of salivary gland components. This study examined the dynamics of salivary microstructure formation, growth, and reorganization using time-lapse imaging over 15 h. Immunofluorescence detection monitored production of individual basement membrane components forming around developing microstructures, and Ki67 assessed proliferation. Dynamic movements in hydrogels were quantified by measuring angular velocity (ω) of rotating salivary microstructures and changes in basement membrane architecture during microstructure growth. Integrin involvement in the dynamic reassembly was assessed using knockdown and inhibitor approaches. Single hS/PCs expanded over 5 days into spherical microstructures typically containing 3–10 cells. In larger macrostructures, proliferation occurred near the peripheral basement membrane that underwent growth-associated cycles of thinning and collapse. De novo secretion of laminin/collagen IV from reorganizing hS/PCs preceded that of perlecan/HSPG2. Microstructures routinely expressed β1 integrin-containing complexes at basement membrane-associated regions and exhibited spontaneous and coordinated rotation during basement membrane maturation. β1 integrin siRNA knockdown at the single-cell state prevented hS/PC microstructure growth. After microstructure formation, β1 integrin knockdown reduced rotation and mean ω by 84%. Blockade of the α1 integrin subunit (CD49a) that associates with β1 reduced mean ω by 66%. Studies presented here show that initial hS/PC structure growth and basement membrane maturation depends on α1β1-integrin mediated signaling. Coordinated cellular motility during neotissue reorganization reminiscent of salivary gland acini was critically dependent both on hS/PC-secretion of laminin,collagen type-IV, and perlecan/HSPG2 and the force-driven interactions of α1β1-integrin activation. We conclude that α1β1-integrin plays a critical role in establishing human salivary gland coordinated structure and function, and that its activation in tissue engineered systems is essential to tissue assembly.
Collapse
Affiliation(s)
- Danielle Wu
- Department of Diagnostic and Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, United States.,Department of BioSciences, Rice University, Houston, TX, United States
| | - Robert L Witt
- Center for Translational Cancer Research, Helen F. Graham Cancer Center & Research Institute, Christiana Care Health Center, Newark, DE, United States.,Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, PA, United States
| | - Daniel A Harrington
- Department of Diagnostic and Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, United States.,Department of BioSciences, Rice University, Houston, TX, United States
| | - Mary C Farach-Carson
- Department of Diagnostic and Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, United States.,Department of BioSciences, Rice University, Houston, TX, United States
| |
Collapse
|
10
|
Koslow M, O'Keefe KJ, Hosseini ZF, Nelson DA, Larsen M. ROCK inhibitor increases proacinar cells in adult salivary gland organoids. Stem Cell Res 2019; 41:101608. [PMID: 31731180 PMCID: PMC7069099 DOI: 10.1016/j.scr.2019.101608] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 09/12/2019] [Accepted: 10/02/2019] [Indexed: 02/07/2023] Open
Abstract
Salisphere-derived adult epithelial cells have been used to improve saliva production of irradiated mouse salivary glands. Importantly, optimization of the cellular composition of salispheres could improve their regenerative capabilities. The Rho Kinase (ROCK) inhibitor, Y27632, has been used to increase the proliferation and reduce apoptosis of progenitor cells grown in vitro. In this study, we investigated whether Y27632 could be used to improve expansion of adult submandibular salivary epithelial progenitor cells or to affect their differentiation potential in different media contexts. Application of Y27632 in medium used previously to grow salispheres promoted expansion of Kit+ and Mist1+ cells, while in simple serum-containing medium Y27632 increased the number of cells that expressed the K5 basal progenitor marker. Salispheres derived from Mist1CreERT2; R26TdTomato mice grown in salisphere media with Y27632 included Mist1-derived cells. When these salispheres were incorporated into 3D organoids, inclusion of Y27632 in the salisphere stage increased the contribution of Mist1-derived cells expressing the proacinar/acinar marker, Aquaporin 5 (AQP5), in response to FGF2-dependent mesenchymal signals. Optimization of the cellular composition of salispheres and organoids can be used to improve the application of adult salivary progenitor cells in regenerative medicine strategies.
Collapse
Affiliation(s)
- Matthew Koslow
- Graduate program in Molecular, Cellular, Developmental and Neural Biology, University at Albany, State University of New York, Albany, NY 12222, USA; Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA; RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
| | - Kevin J O'Keefe
- Graduate program in Molecular, Cellular, Developmental and Neural Biology, University at Albany, State University of New York, Albany, NY 12222, USA; Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA; RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
| | - Zeinab F Hosseini
- Graduate program in Molecular, Cellular, Developmental and Neural Biology, University at Albany, State University of New York, Albany, NY 12222, USA; Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA; RNA Institute, 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; RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA
| | - Melinda Larsen
- Graduate program in Molecular, Cellular, Developmental and Neural Biology, University at Albany, State University of New York, Albany, NY 12222, USA; Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA; RNA Institute, University at Albany, State University of New York, Albany, NY 12222, USA; Department of Biological Sciences, University at Albany, SUNY, 1400 Washington Ave., LSRB 1086, Albany, NY 12222, USA.
| |
Collapse
|
11
|
Abstract
Extracellular matrices (ECMs) are structurally and compositionally diverse networks of collagenous and noncollagenous glycoproteins, glycosaminoglycans, proteoglycans, and associated molecules that together comprise the metazoan matrisome. Proper deposition and assembly of ECM is of profound importance to cell proliferation, survival, and differentiation, and the morphogenesis of tissues and organ systems that define sequential steps in the development of all animals. Importantly, it is now clear that the instructive influence of a particular ECM at various points in development reflects more than a simple summing of component parts; cellular responses also reflect the dynamic assembly and changing topology of embryonic ECM, which in turn affect its biomechanical properties. This review highlights recent advances in understanding how biophysical features attributed to ECM, such as stiffness and viscoelasticity, play important roles in the sculpting of embryonic tissues and the regulation of cell fates. Forces generated within cells and tissues are transmitted both through integrin-based adhesions to ECM, and through cadherin-dependent cell-cell adhesions; the resulting short- and long-range deformations of embryonic tissues drive morphogenesis. This coordinate regulation of cell-ECM and cell-cell adhesive machinery has emerged as a common theme in a variety of developmental processes. In this review we consider select examples in the embryo where ECM is implicated in setting up tissue barriers and boundaries, in resisting pushing or pulling forces, or in constraining or promoting cell and tissue movement. We reflect on how each of these processes contribute to morphogenesis.
Collapse
|
12
|
Pirie E, Ray S, Pan C, Fu W, Powers AF, Polikoff D, Miller CM, Kudrna KM, Harris EN, Lusis AJ, Crooke RM, Lee RG. Mouse genome-wide association studies and systems genetics uncover the genetic architecture associated with hepatic pharmacokinetic and pharmacodynamic properties of a constrained ethyl antisense oligonucleotide targeting Malat1. PLoS Genet 2018; 14:e1007732. [PMID: 30372444 PMCID: PMC6224167 DOI: 10.1371/journal.pgen.1007732] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 11/08/2018] [Accepted: 10/01/2018] [Indexed: 12/31/2022] Open
Abstract
Antisense oligonucleotides (ASOs) have demonstrated variation of efficacy in patient populations. This has prompted our investigation into the contribution of genetic architecture to ASO pharmacokinetics (PK) and pharmacodynamics (PD). Genome wide association (GWA) and transcriptomic analysis in a hybrid mouse diversity panel (HMDP) were used to identify and validate novel genes involved in the uptake and efficacy of a single dose of a Malat1 constrained ethyl (cEt) modified ASO. The GWA of the HMDP identified two significant associations on chromosomes 4 and 10 with hepatic Malat1 ASO concentrations. Stabilin 2 (Stab2) and vesicle associated membrane protein 3 (Vamp3) were identified by cis-eQTL analysis. HMDP strains with lower Stab2 expression and Stab2 KO mice displayed significantly lower PK than strains with higher Stab2 expression and the wild type (WT) animals respectively, confirming the role of Stab2 in regulating hepatic Malat1 ASO uptake. GWA examining ASO efficacy uncovered three loci associated with Malat1 potency: Small Subunit Processome Component (Utp11l) on chromosome 4, Rho associated coiled-coil containing protein kinase 2 (Rock2) and Aci-reductone dioxygenase (Adi1) on chromosome 12. Our results demonstrate the utility of mouse GWAS using the HMDP in detecting genes capable of impacting the uptake of ASOs, and identifies genes critical for the activity of ASOs in vivo.
Collapse
Affiliation(s)
- Elaine Pirie
- Cardiovascular Antisense Drug Discovery Group, Ionis Pharmaceuticals, Carlsbad, California, United States of America
| | - Shayoni Ray
- Cardiovascular Antisense Drug Discovery Group, Ionis Pharmaceuticals, Carlsbad, California, United States of America
| | - Calvin Pan
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Wuxia Fu
- Cardiovascular Antisense Drug Discovery Group, Ionis Pharmaceuticals, Carlsbad, California, United States of America
| | - Andrew F. Powers
- Exploratory Drug Discovery Group, Ionis Pharmaceuticals, Carlsbad, California, United States of America
| | - Danielle Polikoff
- Cardiovascular Antisense Drug Discovery Group, Ionis Pharmaceuticals, Carlsbad, California, United States of America
| | - Colton M. Miller
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Katrina M. Kudrna
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Edward N. Harris
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Aldons J. Lusis
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Rosanne M. Crooke
- Cardiovascular Antisense Drug Discovery Group, Ionis Pharmaceuticals, Carlsbad, California, United States of America
| | - Richard G. Lee
- Cardiovascular Antisense Drug Discovery Group, Ionis Pharmaceuticals, Carlsbad, California, United States of America
| |
Collapse
|
13
|
Kourouklis AP, Nelson CM. Modeling branching morphogenesis using materials with programmable mechanical instabilities. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2018; 6:66-73. [PMID: 30345410 PMCID: PMC6193561 DOI: 10.1016/j.cobme.2018.03.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The architectural features of branching morphogenesis demonstrate exquisite reproducibility among various organs and species despite the unique functionality and biochemical differences of their microenvironment. The regulatory networks that drive branching morphogenesis employ cell-generated and passive mechanical forces, which integrate extracellular signals from the microenvironment into morphogenetic movements. Cell-generated forces function locally to remodel the extracellular matrix (ECM) and control interactions among neighboring cells. Passive mechanical forces are the product of in situ mechanical instabilities that trigger out-of-plane buckling and clefting deformations of adjacent tissues. Many of the molecular and physical signals that underlie buckling and clefting morphogenesis remain unclear and require new experimental strategies to be uncovered. Here, we highlight soft material systems that have been engineered to display programmable buckles and creases. Using synthetic materials to model physicochemical and spatiotemporal features of buckling and clefting morphogenesis might facilitate our understanding of the physical mechanisms that drive branching morphogenesis across different organs and species.
Collapse
Affiliation(s)
- Andreas P. Kourouklis
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544
| | - Celeste M. Nelson
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| |
Collapse
|
14
|
Vining KH, Mooney DJ. Mechanical forces direct stem cell behaviour in development and regeneration. Nat Rev Mol Cell Biol 2017; 18:728-742. [PMID: 29115301 PMCID: PMC5803560 DOI: 10.1038/nrm.2017.108] [Citation(s) in RCA: 984] [Impact Index Per Article: 123.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Stem cells and their local microenvironment, or niche, communicate through mechanical cues to regulate cell fate and cell behaviour and to guide developmental processes. During embryonic development, mechanical forces are involved in patterning and organogenesis. The physical environment of pluripotent stem cells regulates their self-renewal and differentiation. Mechanical and physical cues are also important in adult tissues, where adult stem cells require physical interactions with the extracellular matrix to maintain their potency. In vitro, synthetic models of the stem cell niche can be used to precisely control and manipulate the biophysical and biochemical properties of the stem cell microenvironment and to examine how the mode and magnitude of mechanical cues, such as matrix stiffness or applied forces, direct stem cell differentiation and function. Fundamental insights into the mechanobiology of stem cells also inform the design of artificial niches to support stem cells for regenerative therapies.
Collapse
Affiliation(s)
- Kyle H. Vining
- Wyss Institute for Biologically Inspired Engineering and John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - David J. Mooney
- Wyss Institute for Biologically Inspired Engineering and John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
15
|
Foraida ZI, Kamaldinov T, Nelson DA, Larsen M, Castracane J. Elastin-PLGA hybrid electrospun nanofiber scaffolds for salivary epithelial cell self-organization and polarization. Acta Biomater 2017; 62:116-127. [PMID: 28801269 PMCID: PMC5646366 DOI: 10.1016/j.actbio.2017.08.009] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 07/21/2017] [Accepted: 08/07/2017] [Indexed: 01/24/2023]
Abstract
Development of electrospun nanofibers that mimic the structural, mechanical and biochemical properties of natural extracellular matrices (ECMs) is a promising approach for tissue regeneration. Electrospun fibers of synthetic polymers partially mimic the topography of the ECM, however, their high stiffness, poor hydrophilicity and lack of in vivo-like biochemical cues is not optimal for epithelial cell self-organization and function. In search of a biomimetic scaffold for salivary gland tissue regeneration, we investigated the potential of elastin, an ECM protein, to generate elastin hybrid nanofibers that have favorable physical and biochemical properties for regeneration of the salivary glands. Elastin was introduced to our previously developed poly-lactic-co-glycolic acid (PLGA) nanofiber scaffolds by two methods, blend electrospinning (EP-blend) and covalent conjugation (EP-covalent). Both methods for elastin incorporation into the nanofibers improved the wettability of the scaffolds while only blend electrospinning of elastin-PLGA nanofibers and not surface conjugation of elastin to PLGA fibers, conferred increased elasticity to the nanofibers measured by Young's modulus. After two days, only the blend electrospun nanofiber scaffolds facilitated epithelial cell self-organization into cell clusters, assessed with nuclear area and nearest neighbor distance measurements, leading to the apicobasal polarization of salivary gland epithelial cells after six days, which is vital for cell function. This study suggests that elastin electrospun nanofiber scaffolds have potential application in regenerative therapies for salivary glands and other epithelial organs. STATEMENT OF SIGNIFICANCE Regenerating the salivary glands by mimicking the extracellular matrix (ECM) is a promising approach for long term treatment of salivary gland damage. Despite their topographic similarity to the ECM, electrospun fibers of synthetic polymers lack the biochemical complexity, elasticity and hydrophilicity of the ECM. Elastin is an ECM protein abundant in the salivary glands and responsible for tissue elasticity. Although it's widely used for tissue regeneration of other organs, little is known about its utility in regenerating the salivary tissue. This study describes the use of elastin to improve the elasticity, hydrophilicity and biochemical complexity of synthetic nanofibers and its potential in directing in vivo-like organization of epithelial salivary cells which helps the design of efficient salivary gland regeneration scaffolds.
Collapse
Affiliation(s)
- Zahraa I Foraida
- Colleges of Nanoscale Science and Engineering, SUNY Polytechnic Institute, United States
| | - Tim Kamaldinov
- Colleges of Nanoscale Science and Engineering, SUNY Polytechnic Institute, United States
| | - Deirdre A Nelson
- Department of Biological Sciences, University at Albany, State University of New York, United States
| | - Melinda Larsen
- Department of Biological Sciences, University at Albany, State University of New York, United States.
| | - James Castracane
- Colleges of Nanoscale Science and Engineering, SUNY Polytechnic Institute, United States.
| |
Collapse
|
16
|
Gervais EM, Sequeira SJ, Wang W, Abraham S, Kim JH, Leonard D, DeSantis KA, Larsen M. Par-1b is required for morphogenesis and differentiation of myoepithelial cells during salivary gland development. Organogenesis 2016; 12:194-216. [PMID: 27841695 PMCID: PMC5198941 DOI: 10.1080/15476278.2016.1252887] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/24/2016] [Accepted: 10/21/2016] [Indexed: 01/14/2023] Open
Abstract
The salivary epithelium initiates as a solid mass of epithelial cells that are organized into a primary bud that undergoes morphogenesis and differentiation to yield bilayered acini consisting of interior secretory acinar cells that are surrounded by contractile myoepithelial cells in mature salivary glands. How the primary bud transitions into acini has not been previously documented. We document here that the outer epithelial cells subsequently undergo a vertical compression as they express smooth muscle α-actin and differentiate into myoepithelial cells. The outermost layer of polarized epithelial cells assemble and organize the basal deposition of basement membrane, which requires basal positioning of the polarity protein, Par-1b. Whether Par-1b is required for the vertical compression and differentiation of the myoepithelial cells is unknown. Following manipulation of Par-1b in salivary gland organ explants, Par-1b-inhibited explants showed both a reduced vertical compression of differentiating myoepithelial cells and reduced levels of smooth muscle α-actin. Rac1 knockdown and inhibition of Rac GTPase function also inhibited branching morphogenesis. Since Rac regulates cellular morphology, we investigated a contribution for Rac in myoepithelial cell differentiation. Inhibition of Rac GTPase activity showed a similar reduction in vertical compression and smooth muscle α-actin levels while decreasing the levels of Par-1b protein and altering its basal localization in the outer cells. Inhibition of ROCK, which is required for basal positioning of Par-1b, resulted in mislocalization of Par-1b and loss of vertical cellular compression, but did not significantly alter levels of smooth muscle α-actin in these cells. Overexpression of Par-1b in the presence of Rac inhibition restored basement membrane protein levels and localization. Our results indicate that the basal localization of Par-1b in the outer epithelial cells is required for myoepithelial cell compression, and Par-1b is required for myoepithelial differentiation, regardless of its localization.
Collapse
Affiliation(s)
- Elise M. Gervais
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, USA
- Graduate Program in Molecular, Cellular, Developmental, and Neural Biology, University at Albany, State University of New York, Albany, NY, USA
| | - Sharon J. Sequeira
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, USA
| | - Weihao Wang
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, USA
| | - Stanley Abraham
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, USA
| | - Janice H. Kim
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, USA
| | - Daniel Leonard
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, USA
| | - Kara A. DeSantis
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, USA
- Graduate Program in Molecular, Cellular, Developmental, and Neural Biology, University at Albany, State University of New York, Albany, NY, USA
| | - Melinda Larsen
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, USA
| |
Collapse
|
17
|
Li Y, Kilian KA. Bridging the Gap: From 2D Cell Culture to 3D Microengineered Extracellular Matrices. Adv Healthc Mater 2015; 4:2780-96. [PMID: 26592366 PMCID: PMC4780579 DOI: 10.1002/adhm.201500427] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/05/2015] [Indexed: 12/20/2022]
Abstract
Historically the culture of mammalian cells in the laboratory has been performed on planar substrates with media cocktails that are optimized to maintain phenotype. However, it is becoming increasingly clear that much of biology discerned from 2D studies does not translate well to the 3D microenvironment. Over the last several decades, 2D and 3D microengineering approaches have been developed that better recapitulate the complex architecture and properties of in vivo tissue. Inspired by the infrastructure of the microelectronics industry, lithographic patterning approaches have taken center stage because of the ease in which cell-sized features can be engineered on surfaces and within a broad range of biocompatible materials. Patterning and templating techniques enable precise control over extracellular matrix properties including: composition, mechanics, geometry, cell-cell contact, and diffusion. In this review article we explore how the field of engineered extracellular matrices has evolved with the development of new hydrogel chemistry and the maturation of micro- and nano- fabrication. Guided by the spatiotemporal regulation of cell state in developing tissues, techniques for micropatterning in 2D, pseudo-3D systems, and patterning within 3D hydrogels will be discussed in the context of translating the information gained from 2D systems to synthetic engineered 3D tissues.
Collapse
Affiliation(s)
- Yanfen Li
- Department of Materials Science and Engineering, Department of Bioengineering, Institute for Genomic Biology, Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana IL, 61801
| | - Kristopher A. Kilian
- Department of Materials Science and Engineering, Department of Bioengineering, Institute for Genomic Biology, Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana IL, 61801
| |
Collapse
|
18
|
Chemokine Signaling Controls Integrity of Radial Glial Scaffold in Developing Spinal Cord and Consequential Proper Position of Boundary Cap Cells. J Neurosci 2015; 35:9211-24. [PMID: 26085643 DOI: 10.1523/jneurosci.0156-15.2015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Radial glial cells are the neural progenitors of the developing CNS and have long radial processes that guide radially migrating neurons. The integrity of the radial glial scaffold, in particular proper adhesion between the endfeet of radial processes and the pial basement membrane (BM), is important for the cellular organization of the CNS, as indicated by evidence emerging from the developing cortex. However, the mechanisms underlying the maintenance of radial glial scaffold integrity during development, when the neuroepithelium rapidly expands, are still poorly understood. Here, we addressed this issue in the developing mouse spinal cord. We show that CXCR4, a receptor of chemokine CXCL12, is expressed in spinal cord radial glia. Conditional knock-out of Cxcr4 in radial glia caused disrupted radial glial scaffold with gaps at the pial endfeet layer and consequentially led to an invasion of boundary cap (BC) cells into the spinal cord. Because BC cells are PNS cells normally positioned at the incoming and outgoing axonal roots, their invasion into the spinal cord suggests a compromised CNS/PNS boundary in the absence of CXCL12/CXCR4 signaling. Both disrupted radial glial scaffold and invasion of BC cells into the CNS were also present in mice deficient in CXCR7, a second receptor of CXCL12. We further show that CXCL12 signaling promotes the radial glia adhesion to BM components and activates integrin β1 avidity. Our study unravels a novel molecular mechanism that deploys CXCL12/CXCR4/CXCR7 for the maintenance of radial glial scaffold integrity, which in turn safeguards the CNS/PNS boundary during spinal cord development.
Collapse
|
19
|
Gervais EM, Desantis KA, Pagendarm N, Nelson DA, Enger T, Skarstein K, Liaaen Jensen J, Larsen M. Changes in the Submandibular Salivary Gland Epithelial Cell Subpopulations During Progression of Sjögren's Syndrome-Like Disease in the NOD/ShiLtJ Mouse Model. Anat Rec (Hoboken) 2015; 298:1622-34. [PMID: 26179322 DOI: 10.1002/ar.23190] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Sjögren's syndrome (SS), an autoimmune exocrinopathy, is associated with dysfunction of the secretory salivary gland epithelium, leading to xerostomia. The etiology of SS disease progression is poorly understood as it is typically not diagnosed until late stage. Since mouse models allow the study of disease progression, we investigated the NOD/ShiLtJ mouse to explore temporal changes to the salivary epithelium. In the NOD/ShiLtJ model, SS presents secondary to autoimmune diabetes, and SS disease is reportedly fully established by 20 weeks. We compared epithelial morphology in the submandibular salivary glands (SMG) of NOD/ShiLtJ mice with SMGs from the parental strain at 12, 18, and 22 weeks of age and used immunofluorescence to detect epithelial proteins, including the acinar marker, aquaporin 5, ductal cell marker, cytokeratin 7, myoepithelial cell marker, smooth muscle α-actin, and the basal cell marker, cytokeratin 5, while confirming immune infiltrates with CD45R. We also compared these proteins in the labial salivary glands of human SS patients with control tissues. In the NOD/ShiLtJ SMG, regions of lymphocytic infiltrates were not associated with widespread epithelial tissue degradation; however, there was a decrease in the area of the gland occupied by secretory epithelial cells in favor of ductal epithelial cells. We observed an expansion of cells expressing cytokeratin 5 within the ducts and within the smooth muscle α-actin(+) basal myoepithelial population. The altered acinar/ductal ratio within the NOD/ShiLtJ SMG likely contributes to salivary hypofunction, while the expansion of cytokeratin 5 positive-basal cells may reflect loss of function or indicate a regenerative response.
Collapse
Affiliation(s)
- Elise M Gervais
- Department of Biological Sciences, State University of New York, University at Albany, Albany, New York.,Molecular, Cellular, Developmental, and Neural Biology Graduate Program, State University of New York, University at Albany, Albany, New York
| | - Kara A Desantis
- Department of Biological Sciences, State University of New York, University at Albany, Albany, New York.,Molecular, Cellular, Developmental, and Neural Biology Graduate Program, State University of New York, University at Albany, Albany, New York
| | - Nicholas Pagendarm
- Department of Biological Sciences, State University of New York, University at Albany, Albany, New York
| | - Deirdre A Nelson
- Department of Biological Sciences, State University of New York, University at Albany, Albany, New York
| | - Tone Enger
- Department of Oral Surgery and Oral Medicine, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Kathrine Skarstein
- Department of Clinical Medicine, Section for Pathology, University of Bergen, Bergen, Norway
| | - Janicke Liaaen Jensen
- Department of Oral Surgery and Oral Medicine, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Melinda Larsen
- Department of Biological Sciences, State University of New York, University at Albany, Albany, New York
| |
Collapse
|
20
|
Nelson DA, Larsen M. Heterotypic control of basement membrane dynamics during branching morphogenesis. Dev Biol 2015; 401:103-9. [PMID: 25527075 PMCID: PMC4465071 DOI: 10.1016/j.ydbio.2014.12.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 11/24/2014] [Accepted: 12/09/2014] [Indexed: 02/06/2023]
Abstract
Many mammalian organs undergo branching morphogenesis to create highly arborized structures with maximized surface area for specialized organ function. Cooperative cell-cell and cell-matrix adhesions that sculpt the emerging tissue architecture are guided by dynamic basement membranes. Properties of the basement membrane are reciprocally controlled by the interacting epithelial and mesenchymal cell populations. Here we discuss how basement membrane remodeling is required for branching morphogenesis to regulate cell-matrix and cell-cell adhesions that are required for cell patterning during morphogenesis and how basement membrane impacts morphogenesis by stimulation of cell patterning, force generation, and mechanotransduction. We suggest that in addition to creating mature epithelial architecture, remodeling of the epithelial basement membrane during branching morphogenesis is also essential to promote maturation of the stromal mesenchyme to create mature organ structure. Recapitulation of developmental cell-matrix and cell-cell interactions are of critical importance in tissue engineering and regeneration strategies that seek to restore organ function.
Collapse
Affiliation(s)
- Deirdre A Nelson
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, 1400 Washington Ave, Albany, NY 12222, USA
| | - Melinda Larsen
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, 1400 Washington Ave, Albany, NY 12222, USA.
| |
Collapse
|
21
|
The contribution of specific cell subpopulations to submandibular salivary gland branching morphogenesis. Curr Opin Genet Dev 2015; 32:47-54. [PMID: 25706196 DOI: 10.1016/j.gde.2015.01.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 01/25/2015] [Accepted: 01/27/2015] [Indexed: 11/23/2022]
Abstract
Branching morphogenesis is the developmental program responsible for generating a large surface to volume ratio in many secretory and absorptive organs. To accomplish branching morphogenesis, spatiotemporal regulation of specific cell subpopulations is required. Here, we review recent studies that define the contributions of distinct cell subpopulations to specific cellular processes during branching morphogenesis in the mammalian submandibular salivary gland, including the initiation of the gland, the coordination of cleft formation, and the contribution of stem/progenitor cells to morphogenesis. In conclusion, we provide an overview of technological advances that have opened opportunities to further probe the contributions of specific cell subpopulations and to define the integration of events required for branching morphogenesis.
Collapse
|
22
|
Peters SB, Nelson DA, Kwon HR, Koslow M, DeSantis KA, Larsen M. TGFβ signaling promotes matrix assembly during mechanosensitive embryonic salivary gland restoration. Matrix Biol 2015; 43:109-24. [PMID: 25652203 DOI: 10.1016/j.matbio.2015.01.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 01/25/2015] [Accepted: 01/25/2015] [Indexed: 01/16/2023]
Abstract
Mechanical properties of the microenvironment regulate cell morphology and differentiation within complex organs. However, methods to restore morphogenesis and differentiation in organs in which compliance is suboptimal are poorly understood. We used mechanosensitive mouse salivary gland organ explants grown at different compliance levels together with deoxycholate extraction and immunocytochemistry of the intact, assembled matrices to examine the compliance-dependent assembly and distribution of the extracellular matrix and basement membrane in explants grown at permissive or non-permissive compliance. Extracellular matrix and basement membrane assembly were disrupted in the glands grown at low compliance compared to those grown at high compliance, correlating with defective morphogenesis and decreased myoepithelial cell differentiation. Extracellular matrix and basement membrane assembly as well as myoepithelial differentiation were restored by addition of TGFβ1 and by mechanical rescue, and mechanical rescue was prevented by inhibition of TGFβ signaling during the rescue. We detected a basal accumulation of active integrin β1 in the differentiating myoepithelial cells that formed a continuous peripheral localization around the proacini and in clefts within active sites of morphogenesis in explants that were grown at high compliance. The pattern and levels of integrin β1 activation together with myoepithelial differentiation were interrupted in explants grown at low compliance but were restored upon mechanical rescue or with application of exogenous TGFβ1. These data suggest that therapeutic application of TGFβ1 to tissues disrupted by mechanical signaling should be examined as a method to promote organ remodeling and regeneration.
Collapse
Affiliation(s)
- Sarah B Peters
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, United States
| | - Deirdre A Nelson
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, United States
| | - Hae Ryong Kwon
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, United States; Graduate Program in Molecular, Cellular, Neural, and Developmental Biology, University at Albany, State University of New York, United States
| | - Matthew Koslow
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, United States; Graduate Program in Molecular, Cellular, Neural, and Developmental Biology, University at Albany, State University of New York, United States
| | - Kara A DeSantis
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, United States; Graduate Program in Molecular, Cellular, Neural, and Developmental Biology, University at Albany, State University of New York, United States
| | - Melinda Larsen
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, United States.
| |
Collapse
|
23
|
Bonnans C, Chou J, Werb Z. Remodelling the extracellular matrix in development and disease. Nat Rev Mol Cell Biol 2015; 15:786-801. [PMID: 25415508 DOI: 10.1038/nrm3904] [Citation(s) in RCA: 2864] [Impact Index Per Article: 286.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The extracellular matrix (ECM) is a highly dynamic structure that is present in all tissues and continuously undergoes controlled remodelling. This process involves quantitative and qualitative changes in the ECM, mediated by specific enzymes that are responsible for ECM degradation, such as metalloproteinases. The ECM interacts with cells to regulate diverse functions, including proliferation, migration and differentiation. ECM remodelling is crucial for regulating the morphogenesis of the intestine and lungs, as well as of the mammary and submandibular glands. Dysregulation of ECM composition, structure, stiffness and abundance contributes to several pathological conditions, such as fibrosis and invasive cancer. A better understanding of how the ECM regulates organ structure and function and of how ECM remodelling affects disease progression will contribute to the development of new therapeutics.
Collapse
Affiliation(s)
- Caroline Bonnans
- 1] Department of Anatomy, University of California, 513 Parnassus Avenue, San Francisco, California 94143-0452, USA. [2] Oncology Department, INSERM U661, Functional Genomic Institute, 141 rue de la Cardonille, 34094 Montpellier, France
| | - Jonathan Chou
- 1] Department of Anatomy, University of California, 513 Parnassus Avenue, San Francisco, California 94143-0452, USA. [2] Department of Medicine, University of California, 513 Parnassus Avenue, San Francisco, California 94143-0452, USA
| | - Zena Werb
- Department of Anatomy, University of California, 513 Parnassus Avenue, San Francisco, California 94143-0452, USA
| |
Collapse
|
24
|
Chang KH, Nayak RC, Roy S, Perumbeti A, Wellendorf AM, Bezold KY, Pirman M, Hill SE, Starnes J, Loberg A, Zhou X, Inagami T, Zheng Y, Malik P, Cancelas JA. Vasculopathy-associated hyperangiotensinemia mobilizes haematopoietic stem cells/progenitors through endothelial AT₂R and cytoskeletal dysregulation. Nat Commun 2015; 6:5914. [PMID: 25574809 PMCID: PMC4293039 DOI: 10.1038/ncomms6914] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 11/20/2014] [Indexed: 12/26/2022] Open
Abstract
Patients in organ failure of vascular origin have increased circulating hematopoietic stem cells and progenitors (HSC/P). Plasma levels of angiotensin II (Ang-II), are commonly increased in vasculopathies. Hyperangiotensinemia results in activation of a very distinct Ang-II receptor set, Rho-family GTPase members, and actin in bone marrow endothelial cells (BMEC) and HSC/P, which results in decreased membrane integrin activation in both BMEC and HSC/P, and in HSC/P de-adhesion and mobilization. The Ang-II effect can be reversed pharmacologically and genetically by inhibiting Ang-II production or signaling through BMEC AT2R, HSCP AT1R/AT2R or HSC/P RhoA, but not by interfering with other vascular tone mediators. Hyperangiotensinemia and high counts of circulating HSC/P seen in sickle cell disease (SCD) as a result of vascular damage, is significantly decreased by Ang-II inhibitors. Our data define for the first time the role of Ang-II HSC/P traffic regulation and redefine the hematopoietic consequences of anti-angiotensin therapy in SCD.
Collapse
Affiliation(s)
- Kyung Hee Chang
- 1] Hoxworth Blood Center, University of Cincinnati College of Medicine, 3130 Highland Avenue, Cincinnati, Ohio 45267, USA [2] Division of Experimental Hematology and Cell Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229, USA
| | - Ramesh C Nayak
- Division of Experimental Hematology and Cell Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229, USA
| | - Swarnava Roy
- Division of Experimental Hematology and Cell Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229, USA
| | - Ajay Perumbeti
- Hoxworth Blood Center, University of Cincinnati College of Medicine, 3130 Highland Avenue, Cincinnati, Ohio 45267, USA
| | - Ashley M Wellendorf
- Division of Experimental Hematology and Cell Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229, USA
| | - Katie Y Bezold
- Division of Experimental Hematology and Cell Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229, USA
| | - Megan Pirman
- Hoxworth Blood Center, University of Cincinnati College of Medicine, 3130 Highland Avenue, Cincinnati, Ohio 45267, USA
| | - Sarah E Hill
- Hoxworth Blood Center, University of Cincinnati College of Medicine, 3130 Highland Avenue, Cincinnati, Ohio 45267, USA
| | - Joseph Starnes
- Division of Experimental Hematology and Cell Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229, USA
| | - Anastacia Loberg
- Division of Experimental Hematology and Cell Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229, USA
| | - Xuan Zhou
- Division of Experimental Hematology and Cell Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229, USA
| | - Tadashi Inagami
- Department of Biochemistry, Vanderbilt University School of Medicine, 1161 21st Avenue South, Nashville, Tennessee 37232, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cell Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229, USA
| | - Punam Malik
- Division of Experimental Hematology and Cell Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229, USA
| | - Jose A Cancelas
- 1] Hoxworth Blood Center, University of Cincinnati College of Medicine, 3130 Highland Avenue, Cincinnati, Ohio 45267, USA [2] Division of Experimental Hematology and Cell Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229, USA
| |
Collapse
|
25
|
Bonnans C, Chou J, Werb Z. Remodelling the extracellular matrix in development and disease. Nat Rev Mol Cell Biol 2014; 15:786-801. [PMID: 25415508 DOI: 10.1038/nrm3904.remodelling] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The extracellular matrix (ECM) is a highly dynamic structure that is present in all tissues and continuously undergoes controlled remodelling. This process involves quantitative and qualitative changes in the ECM, mediated by specific enzymes that are responsible for ECM degradation, such as metalloproteinases. The ECM interacts with cells to regulate diverse functions, including proliferation, migration and differentiation. ECM remodelling is crucial for regulating the morphogenesis of the intestine and lungs, as well as of the mammary and submandibular glands. Dysregulation of ECM composition, structure, stiffness and abundance contributes to several pathological conditions, such as fibrosis and invasive cancer. A better understanding of how the ECM regulates organ structure and function and of how ECM remodelling affects disease progression will contribute to the development of new therapeutics.
Collapse
Affiliation(s)
- Caroline Bonnans
- 1] Department of Anatomy, University of California, 513 Parnassus Avenue, San Francisco, California 94143-0452, USA. [2] Oncology Department, INSERM U661, Functional Genomic Institute, 141 rue de la Cardonille, 34094 Montpellier, France
| | - Jonathan Chou
- 1] Department of Anatomy, University of California, 513 Parnassus Avenue, San Francisco, California 94143-0452, USA. [2] Department of Medicine, University of California, 513 Parnassus Avenue, San Francisco, California 94143-0452, USA
| | - Zena Werb
- Department of Anatomy, University of California, 513 Parnassus Avenue, San Francisco, California 94143-0452, USA
| |
Collapse
|
26
|
Abstract
Branching morphogenesis is the developmental program that builds the ramified epithelial trees of various organs, including the airways of the lung, the collecting ducts of the kidney, and the ducts of the mammary and salivary glands. Even though the final geometries of epithelial trees are distinct, the molecular signaling pathways that control branching morphogenesis appear to be conserved across organs and species. However, despite this molecular homology, recent advances in cell lineage analysis and real-time imaging have uncovered surprising differences in the mechanisms that build these diverse tissues. Here, we review these studies and discuss the cellular and physical mechanisms that can contribute to branching morphogenesis.
Collapse
Affiliation(s)
- Victor D Varner
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Celeste M Nelson
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544, USA Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
27
|
Ray S, Fanti JA, Macedo DP, Larsen M. LIM kinase regulation of cytoskeletal dynamics is required for salivary gland branching morphogenesis. Mol Biol Cell 2014; 25:2393-407. [PMID: 24966172 PMCID: PMC4142612 DOI: 10.1091/mbc.e14-02-0705] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
LIMK regulation of actin and microtubule dynamics is required for epithelial regulation of early- and late-stage cleft stabilization and progression. LIMK stimulates focal adhesion assembly and integrin β1 activation in cleft regions, causing fibronectin fibrillogenesis and promoting cleft progression during salivary gland branching morphogenesis. Coordinated actin microfilament and microtubule dynamics is required for salivary gland development, although the mechanisms by which they contribute to branching morphogenesis are not defined. Because LIM kinase (LIMK) regulates both actin and microtubule organization, we investigated the role of LIMK signaling in mouse embryonic submandibular salivary glands using ex vivo organ cultures. Both LIMK 1 and 2 were necessary for branching morphogenesis and functioned to promote epithelial early- and late-stage cleft progression through regulation of both microfilaments and microtubules. LIMK-dependent regulation of these cytoskeletal systems was required to control focal adhesion protein–dependent fibronectin assembly and integrin β1 activation, involving the LIMK effectors cofilin and TPPP/p25, for assembly of the actin- and tubulin-based cytoskeletal systems, respectively. We demonstrate that LIMK regulates the early stages of cleft formation—cleft initiation, stabilization, and progression—via establishment of actin stability. Further, we reveal a novel role for the microtubule assembly factor p25 in regulating stabilization and elongation of late-stage progressing clefts. This study demonstrates the existence of multiple actin- and microtubule-dependent stabilization steps that are controlled by LIMK and are required in cleft progression during branching morphogenesis.
Collapse
Affiliation(s)
- Shayoni Ray
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222
| | - Joseph A Fanti
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222
| | - Diego P Macedo
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222
| | - Melinda Larsen
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222
| |
Collapse
|
28
|
Kinney MA, Hookway TA, Wang Y, McDevitt TC. Engineering three-dimensional stem cell morphogenesis for the development of tissue models and scalable regenerative therapeutics. Ann Biomed Eng 2014; 42:352-67. [PMID: 24297495 PMCID: PMC3939035 DOI: 10.1007/s10439-013-0953-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 11/21/2013] [Indexed: 12/11/2022]
Abstract
The physiochemical stem cell microenvironment regulates the delicate balance between self-renewal and differentiation. The three-dimensional assembly of stem cells facilitates cellular interactions that promote morphogenesis, analogous to the multicellular, heterotypic tissue organization that accompanies embryogenesis. Therefore, expansion and differentiation of stem cells as multicellular aggregates provides a controlled platform for studying the biological and engineering principles underlying spatiotemporal morphogenesis and tissue patterning. Moreover, three-dimensional stem cell cultures are amenable to translational screening applications and therapies, which underscores the broad utility of scalable suspension cultures across laboratory and clinical scales. In this review, we discuss stem cell morphogenesis in the context of fundamental biophysical principles, including the three-dimensional modulation of adhesions, mechanics, and molecular transport and highlight the opportunities to employ stem cell spheroids for tissue modeling, bioprocessing, and regenerative therapies.
Collapse
Affiliation(s)
- Melissa A. Kinney
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology/Emory University, Atlanta, GA, USA
| | - Tracy A. Hookway
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology/Emory University, Atlanta, GA, USA
| | - Yun Wang
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology/Emory University, Atlanta, GA, USA
| | - Todd C. McDevitt
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology/Emory University, Atlanta, GA, USA
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
29
|
Ray S, Yuan D, Dhulekar N, Oztan B, Yener B, Larsen M. Cell-based multi-parametric model of cleft progression during submandibular salivary gland branching morphogenesis. PLoS Comput Biol 2013; 9:e1003319. [PMID: 24277996 PMCID: PMC3836695 DOI: 10.1371/journal.pcbi.1003319] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 09/18/2013] [Indexed: 12/12/2022] Open
Abstract
Cleft formation during submandibular salivary gland branching morphogenesis is the critical step initiating the growth and development of the complex adult organ. Previous experimental studies indicated requirements for several epithelial cellular processes, such as proliferation, migration, cell-cell adhesion, cell-extracellular matrix (matrix) adhesion, and cellular contraction in cleft formation; however, the relative contribution of each of these processes is not fully understood since it is not possible to experimentally manipulate each factor independently. We present here a comprehensive analysis of several cellular parameters regulating cleft progression during branching morphogenesis in the epithelial tissue of an early embryonic salivary gland at a local scale using an on lattice Monte-Carlo simulation model, the Glazier-Graner-Hogeweg model. We utilized measurements from time-lapse images of mouse submandibular gland organ explants to construct a temporally and spatially relevant cell-based 2D model. Our model simulates the effect of cellular proliferation, actomyosin contractility, cell-cell and cell-matrix adhesions on cleft progression, and it was used to test specific hypotheses regarding the function of these parameters in branching morphogenesis. We use innovative features capturing several aspects of cleft morphology and quantitatively analyze clefts formed during functional modification of the cellular parameters. Our simulations predict that a low epithelial mitosis rate and moderate level of actomyosin contractility in the cleft cells promote cleft progression. Raising or lowering levels of contractility and mitosis rate resulted in non-progressive clefts. We also show that lowered cell-cell adhesion in the cleft region and increased cleft cell-matrix adhesions are required for cleft progression. Using a classifier-based analysis, the relative importance of these four contributing cellular factors for effective cleft progression was determined as follows: cleft cell contractility, cleft region cell-cell adhesion strength, epithelial cell mitosis rate, and cell-matrix adhesion strength. Branching morphogenesis is a complex and dynamic embryonic process that creates the structure of many adult organs, including the salivary gland. During this process, many cellular changes occur in the epithelial cells, including changes in cell-cell adhesions, cell-extracellular matrix (matrix) adhesions, cell proliferation, and cellular contraction, resulting in formation of clefts in the epithelial cells of the organ. A comprehensive understanding of the relative contributions of these cellular processes has crucial therapeutic implications for organ regeneration and functional restoration of organ structure in diseased salivary glands. Here, we have developed a cell-based model of cleft progression and simulated cleft progression under conditions of altered cell-cell adhesions, cellular contractility, cell-matrix adhesion and cell proliferation to identify the optimum cellular conditions that cause clefts to progress. The model predicts that cleft progression requires a moderate level of cleft cell contractility, a low epithelial proliferation rate, reduced cell-cell adhesion strength in the cleft and high cell-matrix adhesion strength also in the cleft region. The results of our classification analysis demonstrate that cellular contractility in the cleft cells has a significant effect on cleft progression, followed by cell-cell adhesion strength, rate of cell proliferation, and strength of cell-matrix adhesion energies.
Collapse
Affiliation(s)
- Shayoni Ray
- Department of Biological Sciences, University at Albany, State University of New York, Albany, New York, United States of America
| | - Daniel Yuan
- Department of Computer Science, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Nimit Dhulekar
- Department of Computer Science, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Basak Oztan
- Department of Computer Science, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Bülent Yener
- Department of Computer Science, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Melinda Larsen
- Department of Biological Sciences, University at Albany, State University of New York, Albany, New York, United States of America
- * E-mail:
| |
Collapse
|
30
|
Twiss F, de Rooij J. Cadherin mechanotransduction in tissue remodeling. Cell Mol Life Sci 2013; 70:4101-16. [PMID: 23563964 PMCID: PMC11113614 DOI: 10.1007/s00018-013-1329-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 03/19/2013] [Accepted: 03/19/2013] [Indexed: 12/15/2022]
Abstract
Mechanical forces are increasingly recognized as central factors in the regulation of tissue morphogenesis and homeostasis. Central to the transduction of mechanical information into biochemical signaling is the contractile actomyosin cytoskeleton. Fluctuations in actomyosin contraction are sensed by tension sensitive systems at the interface between actomyosin and cell adhesion complexes. We review the current knowledge about the mechanical coupling of cell-cell junctions to the cytoskeleton and highlight the central role of α-catenin in this linkage. We assemble current knowledge about α-catenin's regulation by tension and about its interactions with a diversity of proteins. We present a model in which α-catenin is a force-regulated platform for a machinery of proteins that orchestrates local cortical remodeling in response to force. Finally, we highlight recently described fundamental processes in tissue morphogenesis and argue where and how this α-catenin-dependent cadherin mechanotransduction may be involved.
Collapse
Affiliation(s)
- Floor Twiss
- Hubrecht Institute for Developmental Biology and Stem Cell Research and University Medical Centre Utrecht, Uppsalalaan 8, 3884 CT, Utrecht, The Netherlands,
| | | |
Collapse
|
31
|
Glukhova MA, Streuli CH. How integrins control breast biology. Curr Opin Cell Biol 2013; 25:633-41. [PMID: 23886475 PMCID: PMC3807876 DOI: 10.1016/j.ceb.2013.06.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 06/28/2013] [Accepted: 06/28/2013] [Indexed: 02/07/2023]
Abstract
This article explores new ideas about how the ECM-integrin axis controls normal and malignant breast biology. We discuss the role of integrins in mammary stem cells, and how cell-matrix interactions regulate ductal and alveolar development and function. We also examine the contribution of integrins to tissue disorganisation and metastasis, and how an altered stromal and ECM tumour microenvironment affects the cancer cell niche both within primary tumours and at distant sites. Finally, we mention novel strategies for integrin-directed breast cancer treatment.
Collapse
Affiliation(s)
- Marina A Glukhova
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom
| | | |
Collapse
|
32
|
Daley WP, Yamada KM. ECM-modulated cellular dynamics as a driving force for tissue morphogenesis. Curr Opin Genet Dev 2013; 23:408-14. [PMID: 23849799 DOI: 10.1016/j.gde.2013.05.005] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 05/16/2013] [Accepted: 05/22/2013] [Indexed: 01/16/2023]
Abstract
The extracellular matrix (ECM) plays diverse regulatory roles throughout development. Coordinate interactions between cells within a tissue and the ECM result in the dynamic remodeling of ECM structure. Both chemical signals and physical forces that result from such microenvironmental remodeling regulate cell behavior that sculpts tissue structure. Here, we review recent discoveries illustrating different ways in which ECM remodeling promotes dynamic cell behavior during tissue morphogenesis. We focus first on new insights that identify localized ECM signaling as a regulator of cell migration, shape, and adhesion during branching morphogenesis. We also review mechanisms by which the ECM and basement membrane can both sculpt and stabilize epithelial tissue structure, using as examples Drosophila egg chamber development and cleft formation in epithelial organs. Finally, we end with an overview of the dynamic mechanisms by which the ECM can regulate stem cell differentiation to contribute to proper tissue morphogenesis.
Collapse
Affiliation(s)
- William P Daley
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States.
| | | |
Collapse
|
33
|
Bentzinger CF, Wang YX, von Maltzahn J, Soleimani VD, Yin H, Rudnicki MA. Fibronectin regulates Wnt7a signaling and satellite cell expansion. Cell Stem Cell 2013; 12:75-87. [PMID: 23290138 DOI: 10.1016/j.stem.2012.09.015] [Citation(s) in RCA: 255] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 06/07/2012] [Accepted: 09/14/2012] [Indexed: 01/07/2023]
Abstract
The influence of the extracellular matrix (ECM) within the stem cell niche remains poorly understood. We found that Syndecan-4 (Sdc4) and Frizzled-7 (Fzd7) form a coreceptor complex in satellite cells and that binding of the ECM glycoprotein Fibronectin (FN) to Sdc4 stimulates the ability of Wnt7a to induce the symmetric expansion of satellite stem cells. Newly activated satellite cells dynamically remodel their niche via transient high-level expression of FN. Knockdown of FN in prospectively isolated satellite cells severely impaired their ability to repopulate the satellite cell niche. Conversely, in vivo overexpression of FN with Wnt7a dramatically stimulated the expansion of satellite stem cells in regenerating muscle. Therefore, activating satellite cells remodel their niche through autologous expression of FN that provides feedback to stimulate Wnt7a signaling through the Fzd7/Sdc4 coreceptor complex. Thus, FN and Wnt7a together regulate the homeostatic levels of satellite stem cells and satellite myogenic cells during regenerative myogenesis.
Collapse
Affiliation(s)
- C Florian Bentzinger
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | | | | | | | | | | |
Collapse
|
34
|
Nelson DA, Manhardt C, Kamath V, Sui Y, Santamaria-Pang A, Can A, Bello M, Corwin A, Dinn SR, Lazare M, Gervais EM, Sequeira SJ, Peters SB, Ginty F, Gerdes MJ, Larsen M. Quantitative single cell analysis of cell population dynamics during submandibular salivary gland development and differentiation. Biol Open 2013; 2:439-47. [PMID: 23789091 PMCID: PMC3654261 DOI: 10.1242/bio.20134309] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 03/27/2013] [Indexed: 12/22/2022] Open
Abstract
Epithelial organ morphogenesis involves reciprocal interactions between epithelial and mesenchymal cell types to balance progenitor cell retention and expansion with cell differentiation for evolution of tissue architecture. Underlying submandibular salivary gland branching morphogenesis is the regulated proliferation and differentiation of perhaps several progenitor cell populations, which have not been characterized throughout development, and yet are critical for understanding organ development, regeneration, and disease. Here we applied a serial multiplexed fluorescent immunohistochemistry technology to map the progressive refinement of the epithelial and mesenchymal cell populations throughout development from embryonic day 14 through postnatal day 20. Using computational single cell analysis methods, we simultaneously mapped the evolving temporal and spatial location of epithelial cells expressing subsets of differentiation and progenitor markers throughout salivary gland development. We mapped epithelial cell differentiation markers, including aquaporin 5, PSP, SABPA, and mucin 10 (acinar cells); cytokeratin 7 (ductal cells); and smooth muscle α-actin (myoepithelial cells) and epithelial progenitor cell markers, cytokeratin 5 and c-kit. We used pairwise correlation and visual mapping of the cells in multiplexed images to quantify the number of single- and double-positive cells expressing these differentiation and progenitor markers at each developmental stage. We identified smooth muscle α-actin as a putative early myoepithelial progenitor marker that is expressed in cytokeratin 5-negative cells. Additionally, our results reveal dynamic expansion and redistributions of c-kit- and K5-positive progenitor cell populations throughout development and in postnatal glands. The data suggest that there are temporally and spatially discreet progenitor populations that contribute to salivary gland development and homeostasis.
Collapse
Affiliation(s)
- Deirdre A Nelson
- Department of Biological Sciences, University at Albany, State University of New York , 1400 Washington Avenue, Albany, NY 12222 , USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Sequeira SJ, Gervais EM, Ray S, Larsen M. Genetic modification and recombination of salivary gland organ cultures. J Vis Exp 2013:e50060. [PMID: 23407326 DOI: 10.3791/50060] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Branching morphogenesis occurs during the development of many organs, and the embryonic mouse submandibular gland (SMG) is a classical model for the study of branching morphogenesis. In the developing SMG, this process involves iterative steps of epithelial bud and duct formation, to ultimately give rise to a complex branched network of acini and ducts, which serve to produce and modify/transport the saliva, respectively, into the oral cavity. The epithelial-associated basement membrane and aspects of the mesenchymal compartment, including the mesenchyme cells, growth factors and the extracellular matrix, produced by these cells, are critical to the branching mechanism, although how the cellular and molecular events are coordinated remains poorly understood. The study of the molecular mechanisms driving epithelial morphogenesis advances our understanding of developmental mechanisms and provides insight into possible regenerative medicine approaches. Such studies have been hampered due to the lack of effective methods for genetic manipulation of the salivary epithelium. Currently, adenoviral transduction represents the most effective method for targeting epithelial cells in adult glands in vivo. However, in embryonic explants, dense mesenchyme and the basement membrane surrounding the epithelial cells impedes viral access to the epithelial cells. If the mesenchyme is removed, the epithelium can be transfected using adenoviruses, and epithelial rudiments can resume branching morphogenesis in the presence of Matrigel or laminin-111. Mesenchyme-free epithelial rudiment growth also requires additional supplementation with soluble growth factors and does not fully recapitulate branching morphogenesis as it occurs in intact glands. Here we describe a technique which facilitates adenoviral transduction of epithelial cells and culture of the transfected epithelium with associated mesenchyme. Following microdissection of the embryonic SMGs, removal of the mesenchyme, and viral infection of the epithelium with a GFP-containing adenovirus, we show that the epithelium spontaneously recombines with uninfected mesenchyme, recapitulating intact SMG glandular structure and branching morphogenesis. The genetically modified epithelial cell population can be easily monitored using standard fluorescence microscopy methods, if fluorescently-tagged adenoviral constructs are used. The tissue recombination method described here is currently the most effective and accessible method for transfection of epithelial cells with a wild-type or mutant vector within a complex 3D tissue construct that does not require generation of transgenic animals.
Collapse
|
36
|
Daley WP, Yamada KM. Cell–ECM Interactions and the Regulation of Epithelial Branching Morphogenesis. EXTRACELLULAR MATRIX IN DEVELOPMENT 2013. [DOI: 10.1007/978-3-642-35935-4_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
37
|
Kim HY, Nelson CM. Extracellular matrix and cytoskeletal dynamics during branching morphogenesis. Organogenesis 2012; 8:56-64. [PMID: 22609561 DOI: 10.4161/org.19813] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Branching morphogenesis is a fundamental developmental process which results in amplification of epithelial surface area for exchanging molecules in organs including the lung, kidney, mammary gland and salivary gland. These complex tree-like structures are built by iterative rounds of simple routines of epithelial morphogenesis, including bud formation, extension, and bifurcation, that require constant remodeling of the extracellular matrix (ECM) and the cytoskeleton. In this review, we highlight the current understanding of the role of the ECM and cytoskeletal dynamics in branching morphogenesis across these different organs. The cellular and molecular mechanisms shared during this morphogenetic process provide insight into the development of other branching organs.
Collapse
Affiliation(s)
- Hye Young Kim
- Department of Chemical and Biological Engineering, Princeton University; Princeton, NJ USA
| | | |
Collapse
|
38
|
Bilgin CC, Ray S, Baydil B, Daley WP, Larsen M, Yener B. Multiscale feature analysis of salivary gland branching morphogenesis. PLoS One 2012; 7:e32906. [PMID: 22403724 PMCID: PMC3293912 DOI: 10.1371/journal.pone.0032906] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 02/07/2012] [Indexed: 11/18/2022] Open
Abstract
Pattern formation in developing tissues involves dynamic spatio-temporal changes in cellular organization and subsequent evolution of functional adult structures. Branching morphogenesis is a developmental mechanism by which patterns are generated in many developing organs, which is controlled by underlying molecular pathways. Understanding the relationship between molecular signaling, cellular behavior and resulting morphological change requires quantification and categorization of the cellular behavior. In this study, tissue-level and cellular changes in developing salivary gland in response to disruption of ROCK-mediated signaling by are modeled by building cell-graphs to compute mathematical features capturing structural properties at multiple scales. These features were used to generate multiscale cell-graph signatures of untreated and ROCK signaling disrupted salivary gland organ explants. From confocal images of mouse submandibular salivary gland organ explants in which epithelial and mesenchymal nuclei were marked, a multiscale feature set capturing global structural properties, local structural properties, spectral, and morphological properties of the tissues was derived. Six feature selection algorithms and multiway modeling of the data was performed to identify distinct subsets of cell graph features that can uniquely classify and differentiate between different cell populations. Multiscale cell-graph analysis was most effective in classification of the tissue state. Cellular and tissue organization, as defined by a multiscale subset of cell-graph features, are both quantitatively distinct in epithelial and mesenchymal cell types both in the presence and absence of ROCK inhibitors. Whereas tensor analysis demonstrate that epithelial tissue was affected the most by inhibition of ROCK signaling, significant multiscale changes in mesenchymal tissue organization were identified with this analysis that were not identified in previous biological studies. We here show how to define and calculate a multiscale feature set as an effective computational approach to identify and quantify changes at multiple biological scales and to distinguish between different states in developing tissues.
Collapse
Affiliation(s)
- Cemal Cagatay Bilgin
- Rensselaer Polytechnic Institute, Computer Science Department, Troy, New York, United States of America
| | - Shayoni Ray
- University at Albany, State University of New York, Department of Biological Sciences, Albany, New York, United States of America
| | - Banu Baydil
- Rensselaer Polytechnic Institute, Computer Science Department, Troy, New York, United States of America
| | - William P. Daley
- University at Albany, State University of New York, Department of Biological Sciences, Albany, New York, United States of America
| | - Melinda Larsen
- University at Albany, State University of New York, Department of Biological Sciences, Albany, New York, United States of America
| | - Bülent Yener
- Rensselaer Polytechnic Institute, Computer Science Department, Troy, New York, United States of America
| |
Collapse
|
39
|
Sequeira SJ, Soscia DA, Oztan B, Mosier AP, Jean-Gilles R, Gadre A, Cady NC, Yener B, Castracane J, Larsen M. The regulation of focal adhesion complex formation and salivary gland epithelial cell organization by nanofibrous PLGA scaffolds. Biomaterials 2012; 33:3175-86. [PMID: 22285464 DOI: 10.1016/j.biomaterials.2012.01.010] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 01/04/2012] [Indexed: 12/31/2022]
Abstract
Nanofiber scaffolds have been useful for engineering tissues derived from mesenchymal cells, but few studies have investigated their applicability for epithelial cell-derived tissues. In this study, we generated nanofiber (250 nm) or microfiber (1200 nm) scaffolds via electrospinning from the polymer, poly-l-lactic-co-glycolic acid (PLGA). Cell-scaffold contacts were visualized using fluorescent immunocytochemistry and laser scanning confocal microscopy. Focal adhesion (FA) proteins, such as phosphorylated FAK (Tyr397), paxillin (Tyr118), talin and vinculin were localized to FA complexes in adult cells grown on planar surfaces but were reduced and diffusely localized in cells grown on nanofiber surfaces, similar to the pattern observed in adult mouse salivary gland tissues. Significant differences in epithelial cell morphology and cell clustering were also observed and quantified, using image segmentation and computational cell-graph analyses. No statistically significant differences in scaffold stiffness between planar PLGA film controls compared to nanofibers scaffolds were detected using nanoindentation with atomic force microscopy, indicating that scaffold topography rather than mechanical properties accounts for changes in cell attachments and cell structure. Finally, PLGA nanofiber scaffolds could support the spontaneous self-organization and branching of dissociated embryonic salivary gland cells. Nanofiber scaffolds may therefore have applicability in the future for engineering an artificial salivary gland.
Collapse
Affiliation(s)
- Sharon J Sequeira
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Ave, LSRB 1086, Albany, NY 12222, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|