1
|
Maden M. An ear for an ear, but only if you are a deomyinid. Proc Natl Acad Sci U S A 2025; 122:e2426545122. [PMID: 39908107 DOI: 10.1073/pnas.2426545122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025] Open
Affiliation(s)
- Malcolm Maden
- Department of Biology, University of Florida, Gainesville, FL 32610
| |
Collapse
|
2
|
Wu W, Daneker M, Turner KT, Jolley MA, Lu L. Identifying Heterogeneous Micromechanical Properties of Biological Tissues via Physics-Informed Neural Networks. SMALL METHODS 2025; 9:e2400620. [PMID: 39091065 PMCID: PMC11747890 DOI: 10.1002/smtd.202400620] [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: 04/29/2024] [Revised: 07/19/2024] [Indexed: 08/04/2024]
Abstract
The heterogeneous micromechanical properties of biological tissues have profound implications across diverse medical and engineering domains. However, identifying full-field heterogeneous elastic properties of soft materials using traditional engineering approaches is fundamentally challenging due to difficulties in estimating local stress fields. Recently, there has been a growing interest in data-driven models for learning full-field mechanical responses, such as displacement and strain, from experimental or synthetic data. However, research studies on inferring full-field elastic properties of materials, a more challenging problem, are scarce, particularly for large deformation, hyperelastic materials. Here, a physics-informed machine learning approach is proposed to identify the elasticity map in nonlinear, large deformation hyperelastic materials. This study reports the prediction accuracies and computational efficiency of physics-informed neural networks (PINNs) in inferring the heterogeneous elasticity maps across materials with structural complexity that closely resemble real tissue microstructure, such as brain, tricuspid valve, and breast cancer tissues. Further, the improved architecture is applied to three hyperelastic constitutive models: Neo-Hookean, Mooney Rivlin, and Gent. The improved network architecture consistently produces accurate estimations of heterogeneous elasticity maps, even when there is up to 10% noise present in the training data.
Collapse
Affiliation(s)
- Wensi Wu
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Division of Cardiology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Mitchell Daneker
- Department of Statistics and Data Science, Yale University, New Haven, CT, 06511, USA
- Department of Chemical and Biochemical Engineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kevin T Turner
- Department of Mechanical Engineering and Applied Mechanics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Matthew A Jolley
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Division of Cardiology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Lu Lu
- Department of Statistics and Data Science, Yale University, New Haven, CT, 06511, USA
- Wu Tsai Institute, Yale University, New Haven, CT, 06510, USA
| |
Collapse
|
3
|
Stewart DC, Brisson BK, Yen WK, Liu Y, Wang C, Ruthel G, Gullberg D, Mauck RL, Maden M, Han L, Volk SW. Type III Collagen Regulates Matrix Architecture and Mechanosensing during Wound Healing. J Invest Dermatol 2024:S0022-202X(24)02078-5. [PMID: 39236902 DOI: 10.1016/j.jid.2024.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 08/05/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024]
Abstract
Postnatal cutaneous wound healing is characterized by development of a collagen-rich scar lacking the architecture and functional integrity of unwounded tissue. Directing cell behaviors to efficiently heal wounds while minimizing scar formation remains a major wound management goal. In this study, we demonstrate type III collagen (COL3) as a critical regulator of re-epithelialization and scar formation during healing of COL3-enriched, regenerative (Acomys), scar-permissive (CD-1 Mus and wild-type Col3B6/B6 mice) and COL3-deficient, scar-promoting (Col3F/F, a murine conditional knockdown model) cutaneous wound models. We define a scar-permissive fibrillar collagen architecture signature characterized by elongated and anisotropically aligned collagen fibers that is dose-dependently suppressed by COL3. Furthermore, loss of COL3 alters how cells interpret their microenvironment-their mechanoperception-such that COL3-deficient cells display mechanically active phenotypes in the absence of increased microenvironmental stiffness through the upregulation and engagement of the profibrotic integrin α11. Further understanding COL3's role in regulating matrix architecture and mechanoresponses may inform clinical strategies that harness proregenerative mechanisms.
Collapse
Affiliation(s)
- Daniel C Stewart
- Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Becky K Brisson
- Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - William K Yen
- Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yuchen Liu
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania, USA
| | - Chao Wang
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania, USA
| | - Gordon Ruthel
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Donald Gullberg
- The Department of Biomedicine, University of Bergen, Bergen, Norway; Centre for Cancer Biomarkers (CCBIO), Norwegian Centre of Excellence, University of Bergen, Bergen, Norway
| | - Robert L Mauck
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA; McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, USA
| | - Malcolm Maden
- Department of Biology, College of Liberal Arts and Sciences, University of Florida, Gainesville, Florida, USA
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania, USA
| | - Susan W Volk
- Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
4
|
Tomasso A, Disela V, Longaker MT, Bartscherer K. Marvels of spiny mouse regeneration: cellular players and their interactions in restoring tissue architecture in mammals. Curr Opin Genet Dev 2024; 87:102228. [PMID: 39047585 DOI: 10.1016/j.gde.2024.102228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/12/2024] [Accepted: 06/30/2024] [Indexed: 07/27/2024]
Abstract
Understanding the cellular and molecular determinants of mammalian tissue regeneration and repair is crucial for developing effective therapies that restore tissue architecture and function. In this review, we focus on the cell types involved in scarless wound response and regeneration of spiny mice (Acomys). Comparative -omics approaches with scar-prone mammals have revealed species-specific peculiarities in cellular behavior during the divergent healing trajectories. We discuss the developing views on which cell types engage in restoring the architecture of spiny mouse tissues through a co-ordinated spatiotemporal response to injury. While yet at the beginning of understanding how cells interact in these fascinating animals to regenerate tissues, spiny mice hold great promise for scar prevention and anti-fibrotic treatments.
Collapse
Affiliation(s)
- Antonio Tomasso
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University - School of Medicine, Department of Surgery, Stanford, CA 94305, USA; Department of Biology/Chemistry, Osnabrück University, Osnabrück 49076, Germany; Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Utrecht 3584CT, the Netherlands. https://twitter.com/@anto_tomasso
| | - Vanessa Disela
- Department of Biology/Chemistry, Osnabrück University, Osnabrück 49076, Germany; Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Utrecht 3584CT, the Netherlands. https://twitter.com/@VDisela
| | - Michael T Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University - School of Medicine, Department of Surgery, Stanford, CA 94305, USA. https://twitter.com/@LongakerLab
| | - Kerstin Bartscherer
- Department of Biology/Chemistry, Osnabrück University, Osnabrück 49076, Germany.
| |
Collapse
|
5
|
Caliaro V, Peurichard D, Chara O. How a reaction-diffusion signal can control spinal cord regeneration in axolotls: A modeling study. iScience 2024; 27:110197. [PMID: 39021793 PMCID: PMC11253152 DOI: 10.1016/j.isci.2024.110197] [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: 09/14/2023] [Revised: 02/07/2024] [Accepted: 06/03/2024] [Indexed: 07/20/2024] Open
Abstract
Axolotls are uniquely able to completely regenerate the spinal cord after amputation. The underlying governing mechanisms of this regenerative response have not yet been fully elucidated. We previously found that spinal cord regeneration is mainly driven by cell-cycle acceleration of ependymal cells, recruited by a hypothetical signal propagating from the injury. However, the nature of the signal and its propagation remain unknown. In this theoretical study, we investigated whether the regeneration-inducing signal can follow a reaction-diffusion process. We developed a computational model, validated it with experimental data, and showed that the signal dynamics can be understood in terms of reaction-diffusion mechanism. By developing a theory of the regenerating outgrowth in the limit of fast reaction-diffusion, we demonstrate that control of regenerative response solely relies on cell-to-signal sensitivity and the signal reaction-diffusion characteristic length. This study lays foundations for further identification of the signal controlling regeneration of the spinal cord.
Collapse
Affiliation(s)
- Valeria Caliaro
- Inria Paris, team MAMBA, Sorbonne Université, CNRS, Université de Paris, Laboratoire Jacques-Louis Lions UMR7598, 75005 Paris, France
| | - Diane Peurichard
- Inria Paris, team MAMBA, Sorbonne Université, CNRS, Université de Paris, Laboratoire Jacques-Louis Lions UMR7598, 75005 Paris, France
| | - Osvaldo Chara
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Nottingham LE12 5RD, UK
- Instituto de Tecnología, Universidad Argentina de la Empresa, Buenos Aires C1073AAO, Argentina
| |
Collapse
|
6
|
Wu W, Daneker M, Turner KT, Jolley MA, Lu L. Identifying heterogeneous micromechanical properties of biological tissues via physics-informed neural networks. ARXIV 2024:arXiv:2402.10741v3. [PMID: 38745694 PMCID: PMC11092874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The heterogeneous micromechanical properties of biological tissues have profound implications across diverse medical and engineering domains. However, identifying full-field heterogeneous elastic properties of soft materials using traditional engineering approaches is fundamentally challenging due to difficulties in estimating local stress fields. Recently, there has been a growing interest in using data-driven models to learn full-field mechanical responses such as displacement and strain from experimental or synthetic data. However, research studies on inferring full-field elastic properties of materials, a more challenging problem, are scarce, particularly for large deformation, hyperelastic materials. Here, we propose a physics-informed machine learning approach to identify the elasticity map in nonlinear, large deformation hyperelastic materials. We evaluate the prediction accuracies and computational efficiency of physics-informed neural networks (PINNs) by inferring the heterogeneous elasticity maps across three materials with structural complexity that closely resemble real tissue patterns, such as brain tissue and tricuspid valve tissue. We further applied our improved architecture to three additional examples of breast cancer tissue and extended our analysis to three hyperelastic constitutive models: Neo-Hookean, Mooney Rivlin, and Gent. Our selected network architecture consistently produced highly accurate estimations of heterogeneous elasticity maps, even when there was up to 10% noise present in the training data.
Collapse
Affiliation(s)
- Wensi Wu
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104
- Division of Cardiology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104
| | - Mitchell Daneker
- Department of Statistics and Data Science, Yale University, New Haven, CT 06511
- Department of Chemical and Biochemical Engineering, University of Pennsylvania, Philadelphia, PA 19104
| | - Kevin T. Turner
- Department of Mechanical Engineering and Applied Mechanics, University of Pennsylvania, Philadelphia, PA 19104
| | - Matthew A. Jolley
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104
- Division of Cardiology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104
| | - Lu Lu
- Department of Statistics and Data Science, Yale University, New Haven, CT 06511
| |
Collapse
|
7
|
Wong D, Martinez J, Quijada P. Exploring the Function of Epicardial Cells Beyond the Surface. Circ Res 2024; 135:353-371. [PMID: 38963865 PMCID: PMC11225799 DOI: 10.1161/circresaha.124.321567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
The epicardium, previously viewed as a passive outer layer around the heart, is now recognized as an essential component in development, regeneration, and repair. In this review, we explore the cellular and molecular makeup of the epicardium, highlighting its roles in heart regeneration and repair in zebrafish and salamanders, as well as its activation in young and adult postnatal mammals. We also examine the latest technologies used to study the function of epicardial cells for therapeutic interventions. Analysis of highly regenerative animal models shows that the epicardium is essential in regulating cardiomyocyte proliferation, transient fibrosis, and neovascularization. However, despite the epicardium's unique cellular programs to resolve cardiac damage, it remains unclear how to replicate these processes in nonregenerative mammalian organisms. During myocardial infarction, epicardial cells secrete signaling factors that modulate fibrotic, vascular, and inflammatory remodeling, which differentially enhance or inhibit cardiac repair. Recent transcriptomic studies have validated the cellular and molecular heterogeneity of the epicardium across various species and developmental stages, shedding further light on its function under pathological conditions. These studies have also provided insights into the function of regulatory epicardial-derived signaling molecules in various diseases, which could lead to new therapies and advances in reparative cardiovascular medicine. Moreover, insights gained from investigating epicardial cell function have initiated the development of novel techniques, including using human pluripotent stem cells and cardiac organoids to model reparative processes within the cardiovascular system. This growing understanding of epicardial function holds the potential for developing innovative therapeutic strategies aimed at addressing developmental heart disorders, enhancing regenerative therapies, and mitigating cardiovascular disease progression.
Collapse
Affiliation(s)
- David Wong
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90029
- Molecular, Cellular and Integrative Physiology Graduate Program, University of California, Los Angeles, CA 90029
| | - Julie Martinez
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90029
- Molecular, Cellular and Integrative Physiology Graduate Program, University of California, Los Angeles, CA 90029
| | - Pearl Quijada
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90029
- Eli and Edythe Broad Stem Research Center, University of California, Los Angeles, CA 90029
- Molecular Biology Institute, University of California, Los Angeles, CA 90029
| |
Collapse
|
8
|
Ramos R, Swedlund B, Ganesan AK, Morsut L, Maini PK, Monuki ES, Lander AD, Chuong CM, Plikus MV. Parsing patterns: Emerging roles of tissue self-organization in health and disease. Cell 2024; 187:3165-3186. [PMID: 38906093 PMCID: PMC11299420 DOI: 10.1016/j.cell.2024.05.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 02/22/2024] [Accepted: 05/08/2024] [Indexed: 06/23/2024]
Abstract
Patterned morphologies, such as segments, spirals, stripes, and spots, frequently emerge during embryogenesis through self-organized coordination between cells. Yet, complex patterns also emerge in adults, suggesting that the capacity for spontaneous self-organization is a ubiquitous property of biological tissues. We review current knowledge on the principles and mechanisms of self-organized patterning in embryonic tissues and explore how these principles and mechanisms apply to adult tissues that exhibit features of patterning. We discuss how and why spontaneous pattern generation is integral to homeostasis and healing of tissues, illustrating it with examples from regenerative biology. We examine how aberrant self-organization underlies diverse pathological states, including inflammatory skin disorders and tumors. Lastly, we posit that based on such blueprints, targeted engineering of pattern-driving molecular circuits can be leveraged for synthetic biology and the generation of organoids with intricate patterns.
Collapse
Affiliation(s)
- Raul Ramos
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
| | - Benjamin Swedlund
- Eli and Edythe Broad CIRM Center, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Anand K Ganesan
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, USA; Department of Dermatology, University of California, Irvine, Irvine, CA, USA
| | - Leonardo Morsut
- Eli and Edythe Broad CIRM Center, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Alfred E. Mann Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Philip K Maini
- Mathematical Institute, University of Oxford, Oxford, UK
| | - Edwin S Monuki
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA; Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA
| | - Arthur D Lander
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, USA.
| | - Cheng-Ming Chuong
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
9
|
Tseng CC, Woolley TE, Jiang TX, Wu P, Maini PK, Widelitz RB, Chuong CM. Gap junctions in Turing-type periodic feather pattern formation. PLoS Biol 2024; 22:e3002636. [PMID: 38743770 PMCID: PMC11161087 DOI: 10.1371/journal.pbio.3002636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/07/2024] [Accepted: 04/22/2024] [Indexed: 05/16/2024] Open
Abstract
Periodic patterning requires coordinated cell-cell interactions at the tissue level. Turing showed, using mathematical modeling, how spatial patterns could arise from the reactions of a diffusive activator-inhibitor pair in an initially homogeneous 2D field. Most activators and inhibitors studied in biological systems are proteins, and the roles of cell-cell interaction, ions, bioelectricity, etc. are only now being identified. Gap junctions (GJs) mediate direct exchanges of ions or small molecules between cells, enabling rapid long-distance communications in a cell collective. They are therefore good candidates for propagating nonprotein-based patterning signals that may act according to the Turing principles. Here, we explore the possible roles of GJs in Turing-type patterning using feather pattern formation as a model. We found 7 of the 12 investigated GJ isoforms are highly dynamically expressed in the developing chicken skin. In ovo functional perturbations of the GJ isoform, connexin 30, by siRNA and the dominant-negative mutant applied before placode development led to disrupted primary feather bud formation. Interestingly, inhibition of gap junctional intercellular communication (GJIC) in the ex vivo skin explant culture allowed the sequential emergence of new feather buds at specific spatial locations relative to the existing primary buds. The results suggest that GJIC may facilitate the propagation of long-distance inhibitory signals. Thus, inhibition of GJs may stimulate Turing-type periodic feather pattern formation during chick skin development, and the removal of GJ activity would enable the emergence of new feather buds if the local environment were competent and the threshold to form buds was reached. We further propose Turing-based computational simulations that can predict the sequential appearance of these ectopic buds. Our models demonstrate how a Turing activator-inhibitor system can continue to generate patterns in the competent morphogenetic field when the level of intercellular communication at the tissue scale is modulated.
Collapse
Affiliation(s)
- Chun-Chih Tseng
- Department of Biochemistry and Molecular Medicine, USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | | | - Ting-Xin Jiang
- Department of Pathology, University of Southern California, Los Angeles, California, United States of America
| | - Ping Wu
- Department of Pathology, University of Southern California, Los Angeles, California, United States of America
| | - Philip K. Maini
- Wolfson Centre for Mathematical Biology, Mathematical Institute, Andrew Wiles Building, University of Oxford, Radcliffe Observatory Quarter, Oxford, United Kingdom
| | - Randall B. Widelitz
- Department of Pathology, University of Southern California, Los Angeles, California, United States of America
| | - Cheng-Ming Chuong
- Department of Pathology, University of Southern California, Los Angeles, California, United States of America
| |
Collapse
|
10
|
Tan CT, Lim CY, Lay K. Modelling Human Hair Follicles-Lessons from Animal Models and Beyond. BIOLOGY 2024; 13:312. [PMID: 38785794 PMCID: PMC11117913 DOI: 10.3390/biology13050312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/26/2024] [Accepted: 04/27/2024] [Indexed: 05/25/2024]
Abstract
The hair follicle is a specialized appendage of the skin that is critical for multiple functions, including thermoregulation, immune surveillance, and sebum production. Mammals are born with a fixed number of hair follicles that develop embryonically. Postnatally, these hair follicles undergo regenerative cycles of regression and growth that recapitulate many of the embryonic signaling pathways. Furthermore, hair cycles have a direct impact on skin regeneration in homeostasis, cutaneous wound healing, and disease conditions such as alopecia. Here, we review the current knowledge of hair follicle formation during embryonic development and the post-natal hair cycle, with an emphasis on the molecular signaling pathways underlying these processes. We then discuss efforts to capitalize on the field's understanding of in vivo mechanisms to bioengineer hair follicles or hair-bearing skin in vitro and how such models may be further improved to develop strategies for hair regeneration.
Collapse
Affiliation(s)
- Chew Teng Tan
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore
| | - Chin Yan Lim
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Kenneth Lay
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore
| |
Collapse
|
11
|
Cotterell A, Griffin M, Downer MA, Parker JB, Wan D, Longaker MT. Understanding wound healing in obesity. World J Exp Med 2024; 14:86898. [PMID: 38590299 PMCID: PMC10999071 DOI: 10.5493/wjem.v14.i1.86898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/30/2023] [Accepted: 01/11/2024] [Indexed: 03/19/2024] Open
Abstract
Obesity has become more prevalent in the global population. It is associated with the development of several diseases including diabetes mellitus, coronary heart disease, and metabolic syndrome. There are a multitude of factors impacted by obesity that may contribute to poor wound healing outcomes. With millions worldwide classified as obese, it is imperative to understand wound healing in these patients. Despite advances in the understanding of wound healing in both healthy and diabetic populations, much is unknown about wound healing in obese patients. This review examines the impact of obesity on wound healing and several animal models that may be used to broaden our understanding in this area. As a growing portion of the population identifies as obese, understanding the underlying mechanisms and how to overcome poor wound healing is of the utmost importance.
Collapse
Affiliation(s)
- Asha Cotterell
- Division of Plastic and Reconstructive Surgery, Stanford University, Palo Alto, CA 94301, United States
| | - Michelle Griffin
- Division of Plastic and Reconstructive Surgery, Stanford University, Palo Alto, CA 94301, United States
| | - Mauricio A Downer
- Stanford University School of Medicine, Stanford University School of Medicine, Palo Alto, CA 94301, United States
| | - Jennifer B Parker
- Stanford University School of Medicine, Stanford University School of Medicine, Palo Alto, CA 94301, United States
| | - Derrick Wan
- Department of Surgery, Stanford University School of Medicine, Hagey Laboratory for Pediatric Regenerative Medicine, Palo Alto, CA 94301, United States
| | - Michael T Longaker
- Department of Surgery, Stanford University School of Medicine, Hagey Laboratory for Pediatric Regenerative Medicine, Palo Alto, CA 94301, United States
| |
Collapse
|
12
|
Kuo CH, Lee GH, Wu HL, Huang JY, Tang MJ. Breaking the symmetry of cell contractility drives tubulogenesis via CXCL1 polarization. Proc Natl Acad Sci U S A 2024; 121:e2315894121. [PMID: 38377213 PMCID: PMC10907267 DOI: 10.1073/pnas.2315894121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/12/2024] [Indexed: 02/22/2024] Open
Abstract
The intricate interplay between biomechanical and biochemical pathways in modulating morphogenesis is an interesting research topic. How biomechanical force regulates epithelial cell tubulogenesis remains poorly understood. Here, we established a model of tubulogenesis by culturing renal proximal tubular epithelial cells on a collagen gel while manipulating contractile force. Epithelial cells were dynamically self-organized into tubule-like structures by augmentation of cell protrusions and cell-cell association. Reduction and asymmetric distribution of phosphorylated myosin light chain 2, the actomyosin contractility, in cells grown on soft matrix preceded tube connection. Notably, reducing matrix stiffness via sonication of collagen fibrils and inhibiting actomyosin contractility with blebbistatin promoted tubulogenesis, whereas inhibition of cytoskeleton polymerization suppressed it. CXC chemokine ligand 1 (CXCL1) expression was transcriptionally upregulated in cells undergoing tubulogenesis. Additionally, inhibiting actomyosin contractility facilitated CXCL1 polarization and cell protrusions preceding tube formation. Conversely, inhibiting the CXCL1-CXC receptor 1 pathway hindered cell protrusions and tubulogenesis. Mechanical property asymmetry with cell-collagen fibril interaction patterns at cell protrusions and along the tube structure supported the association of anisotropic contraction with tube formation. Furthermore, suppressing the mechanosensing machinery of integrin subunit beta 1 reduced CXCL1 expression, collagen remodeling, and impaired tubulogenesis. In summary, symmetry breaking of cell contractility on a soft collagen gel promotes CXCL1 polarization at cell protrusions which in turn facilitates cell-cell association and thus tubule connection.
Collapse
Affiliation(s)
- Cheng-Hsiang Kuo
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan701, Taiwan
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan701, Taiwan
| | - Gang-Hui Lee
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan701, Taiwan
| | - Hua-Lin Wu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan701, Taiwan
| | - Jyun-Yuan Huang
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan701, Taiwan
| | - Ming-Jer Tang
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan701, Taiwan
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan701, Taiwan
| |
Collapse
|
13
|
Kohlhauser M, Tuca A, Kamolz LP. The efficacy of adipose-derived stem cells in burn injuries: a systematic review. Cell Mol Biol Lett 2024; 29:10. [PMID: 38182971 PMCID: PMC10771009 DOI: 10.1186/s11658-023-00526-w] [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: 10/13/2023] [Accepted: 12/18/2023] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND Burn injuries can be associated with prolonged healing, infection, a substantial inflammatory response, extensive scarring, and eventually death. In recent decades, both the mortality rates and long-term survival of severe burn victims have improved significantly, and burn care research has increasingly focused on a better quality of life post-trauma. However, delayed healing, infection, pain and extensive scar formation remain a major challenge in the treatment of burns. ADSCs, a distinct type of mesenchymal stem cells, have been shown to improve the healing process. The aim of this review is to evaluate the efficacy of ADSCs in the treatment of burn injuries. METHODS A systematic review of the literature was conducted using the electronic databases PubMed, Web of Science and Embase. The basic research question was formulated with the PICO framework, whereby the usage of ADSCs in the treatment of burns in vivo was determined as the fundamental inclusion criterion. Additionally, pertinent journals focusing on burns and their treatment were screened manually for eligible studies. The review was registered in PROSPERO and reported according to the PRISMA statement. RESULTS Of the 599 publications screened, 21 were considered relevant to the key question and were included in the present review. The included studies were almost all conducted on rodents, with one exception, where pigs were investigated. 13 of the studies examined the treatment of full-thickness and eight of deep partial-thickness burn injuries. 57,1 percent of the relevant studies have demonstrated that ADSCs exhibit immunomodulatory effects during the inflammatory response. 16 studies have shown improved neovascularisation with the use of ADSCs. 14 studies report positive influences of ADSCs on granulation tissue formation, while 11 studies highlight their efficacy in promoting re-epithelialisation. 11 trials demonstrated an improvement in outcomes during the remodelling phase. CONCLUSION In conclusion, it appears that adipose-derived stem cells demonstrate remarkable efficacy in the field of regenerative medicine. However, the usage of ADSCs in the treatment of burns is still at an early experimental stage, and further investigations are required in order to examine the potential usage of ADSCs in future clinical burn care.
Collapse
Affiliation(s)
- Michael Kohlhauser
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Graz, Austria.
| | - Alexandru Tuca
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
- Department of Surgery, State Hospital Güssing, Güssing, Austria
| | - Lars-Peter Kamolz
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
- COREMED-Cooperative Centre for Regenerative Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, Graz, Austria
| |
Collapse
|
14
|
Lei M, Jiang J, Wang M, Wu W, Zhang J, Liu W, Zhou W, Lai YC, Jiang TX, Widelitz RB, Harn HIC, Yang L, Chuong CM. Epidermal-dermal coupled spheroids are important for tissue pattern regeneration in reconstituted skin explant cultures. NPJ Regen Med 2023; 8:65. [PMID: 37996466 PMCID: PMC10667216 DOI: 10.1038/s41536-023-00340-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 11/09/2023] [Indexed: 11/25/2023] Open
Abstract
Tissue patterning is critical for the development and regeneration of organs. To advance the use of engineered reconstituted skin organs, we study cardinal features important for tissue patterning and hair regeneration. We find they spontaneously form spheroid configurations, with polarized epidermal cells coupled with dermal cells through a newly formed basement membrane. Functionally, the spheroid becomes competent morphogenetic units (CMU) that promote regeneration of tissue patterns. The emergence of new cell types and molecular interactions during CMU formation was analyzed using scRNA-sequencing. Surprisingly, in newborn skin explants, IFNr signaling can induce apical-basal polarity in epidermal cell aggregates. Dermal-Tgfb induces basement membrane formation. Meanwhile, VEGF signaling mediates dermal cell attachment to the epidermal cyst shell, thus forming a CMU. Adult mouse and human fetal scalp cells fail to form a CMU but can be restored by adding IFNr or VEGF to achieve hair regeneration. We find different multi-cellular configurations and molecular pathways are used to achieve morphogenetic competence in developing skin, wound-induced hair neogenesis, and reconstituted explant cultures. Thus, multiple paths can be used to achieve tissue patterning. These insights encourage more studies of "in vitro morphogenesis" which may provide novel strategies to enhance regeneration.
Collapse
Affiliation(s)
- Mingxing Lei
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
- Integrative Stem Cell Center, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan.
| | - Jingwei Jiang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Mengyue Wang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Wang Wu
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Jinwei Zhang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Wanqian Liu
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Wei Zhou
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Yung-Chih Lai
- Integrative Stem Cell Center, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan
| | - Ting-Xin Jiang
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Randall B Widelitz
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Hans I-Chen Harn
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Li Yang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Cheng-Ming Chuong
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
15
|
Lim C, Lim J, Choi S. Wound-Induced Hair Follicle Neogenesis as a Promising Approach for Hair Regeneration. Mol Cells 2023; 46:573-578. [PMID: 37650216 PMCID: PMC10590709 DOI: 10.14348/molcells.2023.0071] [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: 04/29/2023] [Revised: 07/21/2023] [Accepted: 08/16/2023] [Indexed: 09/01/2023] Open
Abstract
The mammalian skin contains hair follicles, which are epidermal appendages that undergo periodic cycles and exhibit mini-organ features, such as discrete stem cell compartments and different cellular components. Wound-induced hair follicle neogenesis (WIHN) is the remarkable ability to regenerate hair follicles after large-scale wounding and occurs in several adult mammals. WIHN is comparable to embryonic hair follicle development in its processes. Researchers are beginning to identify the stem cells that, in response to wounding, develop into neogenic hair follicles, as well as to understand the functions of immune cells, mesenchymal cells, and several signaling pathways that are essential for this process. WIHN represents a promising therapeutic approach to the reprogramming of cellular states for promoting hair follicle regeneration and preventing scar formation. In the scope of this review, we investigate the contribution of several cell types and molecular mechanisms to WIHN.
Collapse
Affiliation(s)
- Chaeryeong Lim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
| | - Jooyoung Lim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
| | - Sekyu Choi
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
- School of Medical Science and Engineering, POSTECH, Pohang 37673, Korea
- School of Interdisciplinary Bioscience and Bioengineering, POSTECH, Pohang 37673, Korea
- Institute for Convergence Research and Education in Advanced Technology (I_CREATE), Yonsei University, Incheon 21983, Korea
| |
Collapse
|
16
|
Thurakkal B, Hari K, Marwaha R, Karki S, Jolly MK, Das T. Collective heterogeneity of mitochondrial potential in contact inhibition of proliferation. Biophys J 2023; 122:3909-3923. [PMID: 37598292 PMCID: PMC10560682 DOI: 10.1016/j.bpj.2023.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 06/05/2023] [Accepted: 08/17/2023] [Indexed: 08/21/2023] Open
Abstract
In the epithelium, cell density and cell proliferation are closely connected to each other through contact inhibition of proliferation (CIP). Depending on cell density, CIP proceeds through three distinct stages: the free-growing stage at low density, the pre-epithelial transition stage at medium density, and the post-epithelial transition stage at high density. Previous studies have elucidated how cell morphology, motion, and mechanics vary in these stages. However, it remains unknown whether cellular metabolism also has a density-dependent behavior. By measuring the mitochondrial membrane potential at different cell densities, here we reveal a heterogeneous landscape of metabolism in the epithelium, which appears qualitatively distinct in three stages of CIP and did not follow the trend of other CIP-associated parameters, which increases or decreases monotonically with increasing cell density. Importantly, epithelial cells established a collective metabolic heterogeneity exclusively in the pre-epithelial transition stage, where the multicellular clusters of high- and low-potential cells emerged. However, in the post-epithelial transition stage, the metabolic potential field became relatively homogeneous. Next, to study the underlying dynamics, we constructed a system biology model, which predicted the role of cell proliferation in metabolic potential toward establishing collective heterogeneity. Further experiments indeed revealed that the metabolic pattern spatially correlated with the proliferation capacity of cells, as measured by the nuclear localization of a pro-proliferation protein, YAP. Finally, experiments perturbing the actomyosin contractility revealed that, while metabolic heterogeneity was maintained in the absence of actomyosin contractility, its ab initio emergence depended on the latter. Taken together, our results revealed a density-dependent collective heterogeneity in the metabolic field of a pre-epithelial transition-stage epithelial monolayer, which may have significant implications for epithelial form and function.
Collapse
Affiliation(s)
- Basil Thurakkal
- Tata Institute of Fundamental Research Hyderabad (TIFR-H), Hyderabad, India
| | - Kishore Hari
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India
| | - Rituraj Marwaha
- Tata Institute of Fundamental Research Hyderabad (TIFR-H), Hyderabad, India
| | - Sanjay Karki
- Tata Institute of Fundamental Research Hyderabad (TIFR-H), Hyderabad, India
| | - Mohit K Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru, India.
| | - Tamal Das
- Tata Institute of Fundamental Research Hyderabad (TIFR-H), Hyderabad, India.
| |
Collapse
|
17
|
Nguyen ED, Fard VN, Kim BY, Collins S, Galey M, Nelson BR, Wakenight P, Gable SM, McKenna A, Bammler TK, MacDonald J, Okamura DM, Shendure J, Beier DR, Ramirez JM, Majesky MW, Millen KJ, Tollis M, Miller DE. Genome Report: chromosome-scale genome assembly of the African spiny mouse (Acomys cahirinus). G3 (BETHESDA, MD.) 2023; 13:jkad177. [PMID: 37552705 PMCID: PMC10542272 DOI: 10.1093/g3journal/jkad177] [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: 04/03/2023] [Revised: 04/03/2023] [Accepted: 06/23/2023] [Indexed: 08/10/2023]
Abstract
There is increasing interest in the African spiny mouse (Acomys cahirinus) as a model organism because of its ability for regeneration of tissue after injury in skin, muscle, and internal organs such as the kidneys. A high-quality reference genome is needed to better understand these regenerative properties at the molecular level. Here, we present an improved reference genome for A. cahirinus generated from long Nanopore sequencing reads. We confirm the quality of our annotations using RNA sequencing data from 4 different tissues. Our genome is of higher contiguity and quality than previously reported genomes from this species and will facilitate ongoing efforts to better understand the regenerative properties of this organism.
Collapse
Affiliation(s)
- Elizabeth Dong Nguyen
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101, USA
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA 98195, USA
| | - Vahid Nikoonejad Fard
- School of Informatics, Computing, and Cyber Systems, Northern Arizona University, Flagstaff, AZ 86011, USA
| | - Bernard Y Kim
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Sarah Collins
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Miranda Galey
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Branden R Nelson
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Paul Wakenight
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Simone M Gable
- School of Informatics, Computing, and Cyber Systems, Northern Arizona University, Flagstaff, AZ 86011, USA
| | - Aaron McKenna
- Department of Molecular & Systems Biology, Dartmouth Geisel School of Medicine, Lebanon, NH 03755, USA
| | - Theo K Bammler
- Department of Environmental & Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA
| | - Jim MacDonald
- Department of Environmental & Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA
| | - Daryl M Okamura
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Jay Shendure
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA 98195, USA
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, Seattle, WA 98195, USA
- Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA 98195, USA
| | - David R Beier
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Jan Marino Ramirez
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA
| | - Mark W Majesky
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101, USA
- Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA 98195, USA
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA 98195, USA
| | - Kathleen J Millen
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA 98195, USA
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Marc Tollis
- School of Informatics, Computing, and Cyber Systems, Northern Arizona University, Flagstaff, AZ 86011, USA
| | - Danny E Miller
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA 98195, USA
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
18
|
Saini K, Cho S, Tewari M, Jalil AR, Wang M, Kasznel AJ, Yamamoto K, Chenoweth DM, Discher DE. Pan-tissue scaling of stiffness versus fibrillar collagen reflects contractility-driven strain that inhibits fibril degradation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.27.559759. [PMID: 37808742 PMCID: PMC10557712 DOI: 10.1101/2023.09.27.559759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Polymer network properties such as stiffness often exhibit characteristic power laws in polymer density and other parameters. However, it remains unclear whether diverse animal tissues, composed of many distinct polymers, exhibit such scaling. Here, we examined many diverse tissues from adult mouse and embryonic chick to determine if stiffness ( E tissue ) follows a power law in relation to the most abundant animal protein, Collagen-I, even with molecular perturbations. We quantified fibrillar collagen in intact tissue by second harmonic generation (SHG) imaging and from tissue extracts by mass spectrometry (MS), and collagenase-mediated decreases were also tracked. Pan-tissue power laws for tissue stiffness versus Collagen-I levels measured by SHG or MS exhibit sub-linear scaling that aligns with results from cellularized gels of Collagen-I but not acellular gels. Inhibition of cellular myosin-II based contraction fits the scaling, and combination with inhibitors of matrix metalloproteinases (MMPs) show collagenase activity is strain - not stress- suppressed in tissues, consistent with past studies of gels and fibrils. Beating embryonic hearts and tendons, which differ in both collagen levels and stiffness by >1000-fold, similarly suppressed collagenases at physiological strains of ∼5%, with fiber-orientation regulating degradation. Scaling of E tissue based on 'use-it-or-lose-it' kinetics provides insight into scaling of organ size, microgravity effects, and regeneration processes while suggesting contractility-driven therapeutics.
Collapse
|
19
|
Avila-Martinez N, Gansevoort M, Verbakel J, Jayaprakash H, Araujo IM, Vitorino M, Tiscornia G, van Kuppevelt TH, Daamen WF. Matrisomal components involved in regenerative wound healing in axolotl and Acomys: implications for biomaterial development. Biomater Sci 2023; 11:6060-6081. [PMID: 37525590 DOI: 10.1039/d3bm00835e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Achieving regeneration in humans has been a long-standing goal of many researchers. Whereas amphibians like the axolotl (Ambystoma mexicanum) are capable of regenerating whole organs and even limbs, most mammals heal their wounds via fibrotic scarring. Recently, the African spiny mouse (Acomys sp.) has been shown to be injury resistant and capable of regenerating several tissue types. A major focal point of research with Acomys has been the identification of drivers of regeneration. In this search, the matrisome components related to the extracellular matrix (ECM) are often overlooked. In this review, we compare Acomys and axolotl skin wound healing and blastema-mediated regeneration by examining their wound healing responses and comparing the expression pattern of matrisome genes, including glycosaminoglycan (GAG) related genes. The goal of this review is to identify matrisome genes that are upregulated during regeneration and could be potential candidates for inclusion in pro-regenerative biomaterials. Research papers describing transcriptomic or proteomic coverage of either skin regeneration or blastema formation in Acomys and axolotl were selected. Matrisome and GAG related genes were extracted from each dataset and the resulting lists of genes were compared. In our analysis, we found several genes that were consistently upregulated, suggesting possible involvement in regenerative processes. Most of the components have been implicated in regulation of cell behavior, extracellular matrix remodeling and wound healing. Incorporation of such pro-regenerative factors into biomaterials may help to shift pro-fibrotic processes to regenerative responses in treated wounds.
Collapse
Affiliation(s)
- Nancy Avila-Martinez
- Department of Medical BioSciences, Radboud Research Institute, Radboud university medical center, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| | - Merel Gansevoort
- Department of Medical BioSciences, Radboud Research Institute, Radboud university medical center, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| | - Juul Verbakel
- Department of Medical BioSciences, Radboud Research Institute, Radboud university medical center, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| | - Haarshaadri Jayaprakash
- Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139, Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, 8005-139, Faro, Portugal
| | - Ines Maria Araujo
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, 8005-139, Faro, Portugal
- Algarve Biomedical Center Research Institute (ABC-RI), University of Algarve, 8005-139, Faro, Portugal
| | - Marta Vitorino
- Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139, Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, 8005-139, Faro, Portugal
- Algarve Biomedical Center Research Institute (ABC-RI), University of Algarve, 8005-139, Faro, Portugal
| | - Gustavo Tiscornia
- Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139, Faro, Portugal
- Eugin Barcelona, Balmes, 236, 08006 Barcelona, Spain
| | - Toin H van Kuppevelt
- Department of Medical BioSciences, Radboud Research Institute, Radboud university medical center, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| | - Willeke F Daamen
- Department of Medical BioSciences, Radboud Research Institute, Radboud university medical center, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| |
Collapse
|
20
|
Huang X, Zhao Y, Liu D, Gu S, Liu Y, Khoong Y, Luo S, Zhang Z, Xia W, Wang M, Liang H, Li M, Li Q, Zan T. ALKBH5-mediated m 6A demethylation fuels cutaneous wound re-epithelialization by enhancing PELI2 mRNA stability. Inflamm Regen 2023; 43:36. [PMID: 37452367 PMCID: PMC10347733 DOI: 10.1186/s41232-023-00288-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 06/20/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND Impaired wound re-epithelialization contributes to cutaneous barrier reconstruction dysfunction. Recently, N6-methyladenosine (m6A) RNA modification has been shown to participate in the determination of RNA fate, and its aberration triggers the pathogenesis of numerous diseases. Howbeit, the function of m6A in wound re-epithelialization remains enigmatic. METHODS Alkbh5‒/‒ mouse was constructed to study the rate of wound re-epithelialization after ALKBH5 ablation. Integrated high-throughput analysis combining methylated RNA immunoprecipitation sequencing (MeRIP-seq) and RNA-seq was used to identify the downstream target of ALKBH5. In vitro and in vivo rescue experiments were conducted to verify the role of the downstream target on the functional phenotype of ALKBH5-deficient cells or animals. Furthermore, the interacting reader protein and regulatory mechanisms were determined through RIP-qPCR, RNA pull-down, and RNA stability assays. RESULTS ALKBH5 was specifically upregulated in the wound edge epidermis. Ablation of ALKBH5 suppressed keratinocyte migration and resulted in delayed wound re-epithelialization in Alkbh5‒/‒ mouse. Integrated high-throughput analysis revealed that PELI2, an E3 ubiquitin protein ligase, serves as the downstream target of ALKBH5. Concordantly, exogenous PELI2 supplementation partially rescued keratinocyte migration and accelerated re-epithelialization in ALKBH5-deficient cells, both in vitro and in vivo. In terms of its mechanism, ALKBH5 promoted PELI2 expression by removing the m6A modification from PELI2 mRNA and enhancing its stability in a YTHDF2-dependent manner. CONCLUSIONS This study identifies ALKBH5 as an endogenous accelerator of wound re-epithelialization, thereby benefiting the development of a reprogrammed m6A targeted therapy for refractory wounds.
Collapse
Affiliation(s)
- Xin Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Yixuan Zhao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China.
| | - Daiming Liu
- Department of Wound Repair, the Second Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Hunan, China
| | - Shuchen Gu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Yunhan Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Yimin Khoong
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Shenying Luo
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Zewei Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Wenzheng Xia
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Meng Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Hsin Liang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Minxiong Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China.
| | - Tao Zan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
21
|
Shellard A, Mayor R. Sculpting with stiffness: rigidity as a regulator of morphogenesis. Biochem Soc Trans 2023; 51:1009-1021. [PMID: 37114613 PMCID: PMC10317161 DOI: 10.1042/bst20220826] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023]
Abstract
From a physical perspective, morphogenesis of tissues results from interplay between their material properties and the mechanical forces exerted on them. The importance of mechanical forces in influencing cell behaviour is widely recognised, whereas the importance of tissue material properties in vivo, like stiffness, has only begun to receive attention in recent years. In this mini-review, we highlight key themes and concepts that have emerged related to how tissue stiffness, a fundamental material property, guides various morphogenetic processes in living organisms.
Collapse
Affiliation(s)
- Adam Shellard
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, U.K
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, U.K
| |
Collapse
|
22
|
Okamura DM, Nguyen ED, Collins SJ, Yoon K, Gere JB, Weiser-Evans MCM, Beier DR, Majesky MW. Mammalian organ regeneration in spiny mice. J Muscle Res Cell Motil 2023; 44:39-52. [PMID: 36131170 DOI: 10.1007/s10974-022-09631-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 08/30/2022] [Indexed: 11/26/2022]
Abstract
Fibrosis-driven solid organ failure is a major world-wide health burden with few therapeutic options. Spiny mice (genus: Acomys) are terrestrial mammals that regenerate severe skin wounds without fibrotic scars to evade predators. Recent studies have shown that spiny mice also regenerate acute ischemic and traumatic injuries to kidney, heart, spinal cord, and skeletal muscle. A common feature of this evolved wound healing response is a lack of formation of fibrotic scar tissue that degrades organ function, inhibits regeneration, and leads to organ failure. Complex tissue regeneration is an extremely rare property among mammalian species. In this article, we discuss the evidence that Acomys represents an emerging model organism that offers a unique opportunity for the biomedical community to investigate and clinically translate molecular mechanisms of scarless wound healing and regeneration of organ function in a mammalian species.
Collapse
Affiliation(s)
- Daryl M Okamura
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Avenue, M/S C9S-5, Seattle, WA, 98101, USA
- Department of Pediatrics, University of Washington, Seattle, WA, 98195, USA
| | - Elizabeth D Nguyen
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Avenue, M/S C9S-5, Seattle, WA, 98101, USA
- Department of Pediatrics, University of Washington, Seattle, WA, 98195, USA
| | - Sarah J Collins
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Avenue, M/S C9S-5, Seattle, WA, 98101, USA
| | - Kevin Yoon
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Avenue, M/S C9S-5, Seattle, WA, 98101, USA
| | - Joshua B Gere
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Avenue, M/S C9S-5, Seattle, WA, 98101, USA
| | - Mary C M Weiser-Evans
- Department of Medicine, Division of Renal Diseases & Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - David R Beier
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Avenue, M/S C9S-5, Seattle, WA, 98101, USA
- Department of Pediatrics, University of Washington, Seattle, WA, 98195, USA
| | - Mark W Majesky
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Avenue, M/S C9S-5, Seattle, WA, 98101, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, 98195, USA.
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, 98195, USA.
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
23
|
Su CY, Liu TY, Wang HV, Yang WC. Histopathological Study on Collagen in Full-Thickness Wound Healing in Fraser's Dolphins ( Lagenodelphis hosei). Animals (Basel) 2023; 13:ani13101681. [PMID: 37238111 DOI: 10.3390/ani13101681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
Fraser's dolphins (Lagenodelphis hosei) possess great healing abilities. Their skin composition can be restored after wounding, including collagen spacing, orientation, and bundle thickness. However, it remains unclear how collagens are involved in the wound-healing process and eventually regain normality in Fraser's dolphins. Learned from the other two scarless healing animals, changes in type III/I collagen composition are believed to modulate the wound healing process and influence the scarring or scarless fate determination in human fetal skin and spiny mouse skin. In the current study, Herovici's, trichrome, and immunofluorescence staining were used on normal and wounded skin samples in Fraser's dolphins. The results suggested that type I collagens were the main type of collagens in the normal skin of Fraser's dolphins, while type III collagens were barely seen. During the wound healing process, type III collagens showed at early wound healing stages, and type I collagen increased in the mature healed wound. In an early healed wound, collagens were organized in a parallel manner, showing a transient hypertrophic-like scar, and eventually restored to normal collagen configuration and adipocyte distribution in the mature healed wound. The remarkable ability to remove excessive collagens merits further investigation to provide new insights into clinical wound management.
Collapse
Affiliation(s)
- Chen-Yi Su
- School of Veterinary Medicine, National Taiwan University, Taipei 106216, Taiwan
| | - Tzu-Yu Liu
- Department of Life Sciences, National Cheng Kung University, Tainan 701, Taiwan
- Marine Biology and Cetacean Research Center, National Cheng Kung University, Tainan 701, Taiwan
| | - Hao-Ven Wang
- Department of Life Sciences, National Cheng Kung University, Tainan 701, Taiwan
- Marine Biology and Cetacean Research Center, National Cheng Kung University, Tainan 701, Taiwan
- Center for Bioscience and Biotechnology, National Cheng Kung University, Tainan 701, Taiwan
| | - Wei-Cheng Yang
- School of Veterinary Medicine, National Taiwan University, Taipei 106216, Taiwan
| |
Collapse
|
24
|
Tomasso A, Koopmans T, Lijnzaad P, Bartscherer K, Seifert AW. An ERK-dependent molecular switch antagonizes fibrosis and promotes regeneration in spiny mice ( Acomys). SCIENCE ADVANCES 2023; 9:eadf2331. [PMID: 37126559 PMCID: PMC10132760 DOI: 10.1126/sciadv.adf2331] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Although most mammals heal injured tissues and organs with scarring, spiny mice (Acomys) naturally regenerate skin and complex musculoskeletal tissues. Now, the core signaling pathways driving mammalian tissue regeneration are poorly characterized. Here, we show that, while immediate extracellular signal-regulated kinase (ERK) activation is a shared feature of scarring (Mus) and regenerating (Acomys) injuries, ERK activity is only sustained at high levels during complex tissue regeneration. Following ERK inhibition, ear punch regeneration in Acomys shifted toward fibrotic repair. Using single-cell RNA sequencing, we identified ERK-responsive cell types. Loss- and gain-of-function experiments prompted us to uncover fibroblast growth factor and ErbB signaling as upstream ERK regulators of regeneration. The ectopic activation of ERK in scar-prone injuries induced a pro-regenerative response, including cell proliferation, extracellular matrix remodeling, and hair follicle neogenesis. Our data detail an important distinction in ERK activity between regenerating and poorly regenerating adult mammals and open avenues to redirect fibrotic repair toward regenerative healing.
Collapse
Affiliation(s)
- Antonio Tomasso
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, Münster 48149, Germany
- Cells in Motion Cluster of Excellence-International Max Planck Research School (CiM-IMPRS Graduate Program), Münster 48149, Germany
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Uppsalalaan 8, Utrecht 3584CT, Netherlands
- Department of Biology/Chemistry, Osnabrück University, Barbarastrasse 11, Osnabrück 49076, Germany
- Department of Biology, University of Kentucky, 101 T.H. Morgan Building, Lexington, KY 40506, USA
| | - Tim Koopmans
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Uppsalalaan 8, Utrecht 3584CT, Netherlands
- Department of Biology/Chemistry, Osnabrück University, Barbarastrasse 11, Osnabrück 49076, Germany
| | - Philip Lijnzaad
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, Netherlands
| | - Kerstin Bartscherer
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, Münster 48149, Germany
- Cells in Motion Cluster of Excellence-International Max Planck Research School (CiM-IMPRS Graduate Program), Münster 48149, Germany
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Uppsalalaan 8, Utrecht 3584CT, Netherlands
- Department of Biology/Chemistry, Osnabrück University, Barbarastrasse 11, Osnabrück 49076, Germany
| | - Ashley W Seifert
- Department of Biology, University of Kentucky, 101 T.H. Morgan Building, Lexington, KY 40506, USA
| |
Collapse
|
25
|
Tseng CC, Woolley TE, Jiang TX, Wu P, Maini PK, Widelitz RB, Chuong CM. Gap junctions in Turing-type periodic feather pattern formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.15.537019. [PMID: 37090608 PMCID: PMC10120740 DOI: 10.1101/2023.04.15.537019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Periodic patterning requires coordinated cell-cell interactions at the tissue level. Turing showed, using mathematical modeling, how spatial patterns could arise from the reactions of a diffusive activator-inhibitor pair in an initially homogenous two-dimensional field. Most activators and inhibitors studied in biological systems are proteins, and the roles of cell-cell interaction, ions, bioelectricity, etc. are only now being identified. Gap junctions (GJs) mediate direct exchanges of ions or small molecules between cells, enabling rapid long-distance communications in a cell collective. They are therefore good candidates for propagating non-protein-based patterning signals that may act according to the Turing principles. Here, we explore the possible roles of GJs in Turing-type patterning using feather pattern formation as a model. We found seven of the twelve investigated GJ isoforms are highly dynamically expressed in the developing chicken skin. In ovo functional perturbations of the GJ isoform, connexin 30, by siRNA and the dominant-negative mutant applied before placode development led to disrupted primary feather bud formation, including patches of smooth skin and buds of irregular sizes. Later, after the primary feather arrays were laid out, inhibition of gap junctional intercellular communication in the ex vivo skin explant culture allowed the emergence of new feather buds in temporal waves at specific spatial locations relative to the existing primary buds. The results suggest that gap junctional communication may facilitate the propagation of long-distance inhibitory signals. Thus, the removal of GJ activity would enable the emergence of new feather buds if the local environment is competent and the threshold to form buds is reached. We propose Turing-based computational simulations that can predict the appearance of these ectopic bud waves. Our models demonstrate how a Turing activator-inhibitor system can continue to generate patterns in the competent morphogenetic field when the level of intercellular communication at the tissue scale is modulated.
Collapse
Affiliation(s)
- Chun-Chih Tseng
- Department of Biochemistry and Molecular Medicine, USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, U.S.A
- Current address: Department of Molecular Biology, Princeton University, Princeton, NJ 08540, U.S.A
| | - Thomas E. Woolley
- School of Mathematics, Cardiff University, Senghennydd Road, Cardiff, CF24 4AG, U.K
| | - Ting-Xin Jiang
- Department of Pathology, University of Southern California, Los Angeles, CA 90033, U.S.A
| | - Ping Wu
- Department of Pathology, University of Southern California, Los Angeles, CA 90033, U.S.A
| | - Philip K. Maini
- Wolfson Centre for Mathematical Biology, Mathematical Institute, Andrew Wiles Building, University of Oxford, Radcliffe Observatory Quarter, Woodstock Road, Oxford OX2 6GG, U.K
| | - Randall B. Widelitz
- Department of Pathology, University of Southern California, Los Angeles, CA 90033, U.S.A
| | - Cheng-Ming Chuong
- Department of Pathology, University of Southern California, Los Angeles, CA 90033, U.S.A
| |
Collapse
|
26
|
Nguyen ED, Fard VN, Kim BY, Collins S, Galey M, Nelson BR, Wakenight P, Gable SM, McKenna A, Bammler TK, MacDonald J, Okamura DM, Shendure J, Beier DR, Ramirez JM, Majesky MW, Millen KJ, Tollis M, Miller DE. GENOME REPORT: Chromosome-scale genome assembly of the African spiny mouse ( Acomys cahirinus ). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.03.535372. [PMID: 37066261 PMCID: PMC10103962 DOI: 10.1101/2023.04.03.535372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
There is increasing interest in the African spiny mouse ( Acomys cahirinus ) as a model organism because of its ability for regeneration of tissue after injury in skin, muscle, and internal organs such as the kidneys. A high-quality reference genome is needed to better understand these regenerative properties at the molecular level. Here, we present an improved reference genome for A. cahirinus generated from long Nanopore sequencing reads. We confirm the quality of our annotations using RNA sequencing data from four different tissues. Our genome is of higher contiguity and quality than previously reported genomes from this species and will facilitate ongoing efforts to better understand the regenerative properties of this organism.
Collapse
Affiliation(s)
- Elizabeth Dong Nguyen
- Department of Pediatrics, University of Washington, Seattle, WA
- Center for Developmental Biology & Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA
| | - Vahid Nikoonejad Fard
- School of Informatics, Computing, and Cyber Systems, Northern Arizona University, Flagstaff, AZ
| | - Bernard Y. Kim
- Department of Biology, Stanford University, Stanford, CA
| | - Sarah Collins
- Center for Developmental Biology & Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA
| | - Miranda Galey
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, WA
| | - Branden R. Nelson
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA
| | - Paul Wakenight
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA
| | - Simone M. Gable
- School of Informatics, Computing, and Cyber Systems, Northern Arizona University, Flagstaff, AZ
| | - Aaron McKenna
- Department of Molecular & Systems Biology, Dartmouth Geisel School of Medicine, Lebanon, NH
| | - Theo K. Bammler
- Department of Environmental & Occupational Health Sciences, University of Washington, Seattle, WA
| | - Jim MacDonald
- Department of Environmental & Occupational Health Sciences, University of Washington, Seattle, WA
| | - Daryl M. Okamura
- Department of Pediatrics, University of Washington, Seattle, WA
- Center for Developmental Biology & Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA
| | - Jay Shendure
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA
- Department of Genome Sciences, University of Washington, Seattle, WA
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA
- Howard Hughes Medical Institute, Seattle, WA
- Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA
| | - David R. Beier
- Department of Pediatrics, University of Washington, Seattle, WA
- Center for Developmental Biology & Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA
| | - Jan Marino Ramirez
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA
- Department of Neurological Surgery, University of Washington, Seattle WA
| | - Mark W. Majesky
- Department of Pediatrics, University of Washington, Seattle, WA
- Center for Developmental Biology & Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA
- Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA
| | - Kathleen J. Millen
- Department of Pediatrics, University of Washington, Seattle, WA
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA
| | - Marc Tollis
- School of Informatics, Computing, and Cyber Systems, Northern Arizona University, Flagstaff, AZ
| | - Danny E. Miller
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, WA
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA
| |
Collapse
|
27
|
Oak ASW, Cotsarelis G. Wound-Induced Hair Neogenesis: A Portal to the Development of New Therapies for Hair Loss and Wound Regeneration. Cold Spring Harb Perspect Biol 2023; 15:a041239. [PMID: 36123030 PMCID: PMC9899649 DOI: 10.1101/cshperspect.a041239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Adult mammals retain the remarkable ability to regenerate hair follicles after wounding. Wound-induced hair neogenesis (WIHN) in many ways recapitulates embryogenesis. The origin of the stem cells that give rise to a nascent hair follicle after wounding and the role of mesenchymal cells and signaling pathways responsible for this regenerative phenomenon are slowly being elucidated. WIHN provides a potential therapeutic window for manipulating cell fate by the introduction of factors during the wound healing process to enhance hair follicle formation.
Collapse
Affiliation(s)
- Allen S W Oak
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - George Cotsarelis
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
28
|
Abstract
The efficacy of implanted biomaterials is largely dependent on the response of the host's immune and stromal cells. Severe foreign body response (FBR) can impede the integration of the implant into the host tissue and compromise the intended mechanical and biochemical function. Many features of FBR, including late-stage fibrotic encapsulation of implants, parallel the formation of fibrotic scar tissue after tissue injury. Regenerative organisms like zebrafish and salamanders can avoid fibrosis after injury entirely, but FBR in these research organisms is rarely investigated because their immune competence is much lower than humans. The recent characterization of a regenerative mammal, the spiny mouse (Acomys), has inspired us to take a closer look at cellular regulation in regenerative organisms across the animal kingdom for insights into avoiding FBR in humans. Here, we highlight how major features of regeneration, such as blastema formation, macrophage polarization, and matrix composition, can be modulated across a range of regenerative research organisms to elucidate common features that may be harnessed to minimize FBR. Leveraging a deeper understanding of regenerative biology for biomaterial design may help to reduce FBR and improve device integration and performance.
Collapse
Affiliation(s)
- Sunaina Sapru
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Michele N Dill
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Chelsey S Simmons
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, Florida 32611, United States.,J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, United States
| |
Collapse
|
29
|
Xue Y, Lim CH, Plikus MV, Ito M, Cotsarelis G, Garza LA. Wound-Induced Hair Neogenesis Model. J Invest Dermatol 2022; 142:2565-2569. [PMID: 36153062 PMCID: PMC9549415 DOI: 10.1016/j.jid.2022.07.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 11/27/2022]
Abstract
Skin wounds in adult mammals typically heal with a fibrotic scar and fail to restore ectodermal appendages, such as hair follicles or adipose tissue. Intriguingly, new hair follicles regenerate in the center of large full-thickness wounds of mice in a process called wound-induced hair neogenesis (WIHN). WIHN is followed by neogenesis of dermal adipose tissue. Both neogenic events reactivate embryonic-like cellular and molecular programs. The WIHN model provides a platform for studying mammalian regeneration, and findings from this model could instruct future regenerative medicine interventions for treating wounds and alopecia. Since Ito et al. rediscovered WIHN 15 years ago, numerous investigators have worked on the WIHN model using varying wounding protocols and model interpretations. Because a variety of factors, including environmental variables and choice of mouse strains, can affect the outcomes of a WIHN study, the purpose of this article is to provide an overview of the experimental variables that impact WIHN so that experiments between laboratories can be compared in a meaningful manner.
Collapse
Affiliation(s)
- Yingchao Xue
- Department of Dermatology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Chae Ho Lim
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, New York, USA; Department of Cell Biology, New York University School of Medicine, New York, New York, USA
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California, Irvine, Irvine, California, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, California, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, California, USA
| | - Mayumi Ito
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, New York, USA; Department of Cell Biology, New York University School of Medicine, New York, New York, USA
| | - George Cotsarelis
- Kligman Laboratories, Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Luis A Garza
- Department of Dermatology, Johns Hopkins University, Baltimore, Maryland, USA; Department of Cell Biology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA; Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA.
| |
Collapse
|
30
|
Topological Distribution of Wound Stiffness Modulates Wound-Induced Hair Follicle Neogenesis. Pharmaceutics 2022; 14:pharmaceutics14091926. [PMID: 36145674 PMCID: PMC9504897 DOI: 10.3390/pharmaceutics14091926] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/02/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
In the large full-thickness mouse skin regeneration model, wound-induced hair neogenesis (WIHN) occurs in the wound center. This implies a spatial regulation of hair regeneration. The role of mechanotransduction during tissue regeneration is poorly understood. Here, we created wounds with equal area but different shapes to understand if perturbing mechanical forces change the area and quantity of de novo hair regeneration. Atomic force microscopy of wound stiffness demonstrated a stiffness gradient across the wound with the wound center softer than the margin. Reducing mechanotransduction signals using FAK or myosin II inhibitors significantly increased WIHN and, conversely, enhancing these signals with an actin stabilizer reduced WIHN. Here, α-SMA was downregulated in FAK inhibitor-treated wounds and lowered wound stiffness. Wound center epithelial cells exhibited a spherical morphology relative to wound margin cells. Differential gene expression analysis of FAK inhibitor-treated wound RNAseq data showed that cytoskeleton-, integrin-, and matrix-associated genes were downregulated, while hair follicular neogenesis, cell proliferation, and cell signaling genes were upregulated. Immunohistochemistry staining showed that FAK inhibition increased pSTAT3 nuclear staining in the regenerative wound center, implying enhanced signaling for hair follicular neogenesis. These findings suggest that controlling wound stiffness modulates tissue regeneration encompassing epithelial competence, tissue patterning, and regeneration during wound healing.
Collapse
|
31
|
Jurickova I, Bonkowski E, Angerman E, Novak E, Huron A, Akers, G, Iwasawa K, Braun T, Hadar R, Hooker M, Han S, Cutler DJ, Okou DT, Kugathasan S, Jegga A, Wells J, Takebe T, Mollen KP, Haberman Y, Denson LA. Eicosatetraynoic Acid and Butyrate Regulate Human Intestinal Organoid Mitochondrial and Extracellular Matrix Pathways Implicated in Crohn's Disease Strictures. Inflamm Bowel Dis 2022; 28:988-1003. [PMID: 35259271 PMCID: PMC9247849 DOI: 10.1093/ibd/izac037] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Indexed: 12/19/2022]
Abstract
BACKGROUND Perturbagen analysis of Crohn's disease (CD) ileal gene expression data identified small molecules including eicosatetraynoic acid (ETYA), which may exert an antifibrotic effect. We developed a patient-specific human intestinal organoid (HIO) model system to test small molecule regulation of mitochondrial and wound-healing functions implicated in stricturing behavior. METHODS HIOs were made from CD induced pluripotent stem cells with and without a loss-of-function haplotype in the DUOX2 gene implicated in ileal homeostasis and characterized under basal conditions and following exposure to butyrate and ETYA using RNA sequencing, flow cytometry, and immunofluorescent and polarized light microscopy. Mitochondrial activity was measured using high-resolution respirometry and tissue stiffness using atomic force microscopy. RESULTS HIOs expressed core mitochondrial and extracellular matrix (ECM) genes and enriched biologic functions implicated in CD ileal strictures; ECM gene expression was suppressed by both butyrate and ETYA, with butyrate also suppressing genes regulating epithelial proliferation. Consistent with this, butyrate, but not ETYA, exerted a profound effect on HIO epithelial mitochondrial function, reactive oxygen species production, and cellular abundance. Butyrate and ETYA suppressed HIO expression of alpha smooth muscle actin expressed by myofibroblasts, type I collagen, and collagen protein abundance. HIOs exhibited tissue stiffness comparable to normal human ileum; this was reduced by chronic ETYA exposure in HIOs carrying the DUOX2 loss-of-function haplotype. CONCLUSIONS ETYA regulates ECM genes implicated in strictures and suppresses collagen content and tissue stiffness in an HIO model. HIOs provide a platform to test personalized therapeutics, including small molecules prioritized by perturbagen analysis.
Collapse
Affiliation(s)
- Ingrid Jurickova
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Erin Bonkowski
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Elizabeth Angerman
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Elizabeth Novak
- Division of General and Thoracic Surgery, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Alex Huron
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Grayce Akers,
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kentaro Iwasawa
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, the University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Biomedical Informatics, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Tzipi Braun
- Department of Pediatrics, Sheba Medical Center, Tel-Aviv University, Tel-HaShomer, Israel
| | - Rotem Hadar
- Department of Pediatrics, Sheba Medical Center, Tel-Aviv University, Tel-HaShomer, Israel
| | - Maria Hooker
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Sarah Han
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - David J Cutler
- Department of Human Genetics, Emory University, Atlanta, GA, USA
| | - David T Okou
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Subra Kugathasan
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Anil Jegga
- Division of Biomedical Informatics, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - James Wells
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Takanori Takebe
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, the University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Institute of Research, Tokyo Medical and Dental University, Japan
| | - Kevin P Mollen
- Division of General and Thoracic Surgery, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yael Haberman
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, the University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Pediatrics, Sheba Medical Center, Tel-Aviv University, Tel-HaShomer, Israel
| | - Lee A Denson
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
32
|
Lim CH, Ito M. Niche stiffness regulates stem cell aging. NATURE AGING 2022; 2:568-569. [PMID: 37117779 DOI: 10.1038/s43587-022-00259-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Affiliation(s)
- Chae Ho Lim
- The Ronald O. Perelman Department of Dermatology, New York University, School of Medicine, New York, NY, USA.
- Department of Cell Biology, New York University, School of Medicine, New York, NY, USA.
| | - Mayumi Ito
- The Ronald O. Perelman Department of Dermatology, New York University, School of Medicine, New York, NY, USA.
- Department of Cell Biology, New York University, School of Medicine, New York, NY, USA.
| |
Collapse
|
33
|
Xue Y, Reddy SK, Garza LA. Toward Understanding Wound Immunology for High-Fidelity Skin Regeneration. Cold Spring Harb Perspect Biol 2022; 14:a041241. [PMID: 35667792 PMCID: PMC9248820 DOI: 10.1101/cshperspect.a041241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Effective tissue repair is vital for the survival of organisms. Yet, how the immune system coordinates with tissue stem cells (SCs) to effect postnatal tissue restoration remains elusive. This review presents current knowledge surrounding wound-induced SC and immune signaling that favors tissue repair, including wound healing and regeneration. We discuss factors that affect regenerative capacities among organisms and the dynamics of local immune cells and SCs during reepithelialization. We also present recent insights into how immune niches communicate with SCs or other body systems to restore the epithelial architecture. Additionally, we summarize our findings on functional wound regeneration, specifically how alarmin (double-stranded RNA [dsRNA])-activated Toll-like receptor signaling and host-microbe interaction-related immune pathways alter the regenerative property of skin SCs. Last, we touch on mechanisms by which known immunologic cellular and molecular signaling might boost the skin's regenerative property. Overall, this review will provide insights into how therapeutically modulating immune signaling could enhance postnatal tissue regeneration.
Collapse
Affiliation(s)
| | - Sashank K Reddy
- Department of Plastic and Reconstructive Surgery
- Department of Biomedical Engineering
- Institute for NanoBioTechnology
| | - Luis A Garza
- Department of Dermatology
- Department of Cell Biology
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland 21231, USA
| |
Collapse
|
34
|
Riquelme-Guzmán C, Beck T, Edwards-Jorquera S, Schlüßler R, Müller P, Guck J, Möllmert S, Sandoval-Guzmán T. In vivo assessment of mechanical properties during axolotl development and regeneration using confocal Brillouin microscopy. Open Biol 2022; 12:220078. [PMID: 35728623 PMCID: PMC9213112 DOI: 10.1098/rsob.220078] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
In processes such as development and regeneration, where large cellular and tissue rearrangements occur, cell fate and behaviour are strongly influenced by tissue mechanics. While most well-established tools probing mechanical properties require an invasive sample preparation, confocal Brillouin microscopy captures mechanical parameters optically with high resolution in a contact-free and label-free fashion. In this work, we took advantage of this tool and the transparency of the highly regenerative axolotl to probe its mechanical properties in vivo for the first time. We mapped the Brillouin frequency shift with high resolution in developing limbs and regenerating digits, the most studied structures in the axolotl. We detected a gradual increase in the cartilage Brillouin frequency shift, suggesting decreasing tissue compressibility during both development and regeneration. Moreover, we were able to correlate such an increase with the regeneration stage, which was undetected with fluorescence microscopy imaging. The present work evidences the potential of Brillouin microscopy to unravel the mechanical changes occurring in vivo in axolotls, setting the basis to apply this technique in the growing field of epimorphic regeneration.
Collapse
Affiliation(s)
- Camilo Riquelme-Guzmán
- CRTD/Center for Regenerative Therapies TU Dresden, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany,Department of Internal Medicine 3, Center for Healthy Aging, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Timon Beck
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany,Max Planck Institute for the Science of Light and Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Sandra Edwards-Jorquera
- Department of Internal Medicine 3, Center for Healthy Aging, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Raimund Schlüßler
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Paul Müller
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany,Max Planck Institute for the Science of Light and Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Jochen Guck
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany,Max Planck Institute for the Science of Light and Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Stephanie Möllmert
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany,Max Planck Institute for the Science of Light and Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Tatiana Sandoval-Guzmán
- Department of Internal Medicine 3, Center for Healthy Aging, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany,Paul Langerhans Institute Dresden, Helmholtz Centre Munich, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
35
|
Direct Reprograming of Mouse Fibroblasts into Dermal Papilla Cells via Small Molecules. Int J Mol Sci 2022; 23:ijms23084213. [PMID: 35457029 PMCID: PMC9030401 DOI: 10.3390/ijms23084213] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 03/29/2022] [Accepted: 04/07/2022] [Indexed: 01/27/2023] Open
Abstract
The reprogramming of somatic fibroblasts into alternative cell linages could provide a promising source of cells for regenerative medicine and cell therapy. However, the direct conversion of fibroblasts into other functional cell types is still challenging. In this study, we show that dermal-papilla-cell-like cells (DPC-LCs) can be generated by treating fibroblasts, including L929 mouse fibroblast cell lines and somatic mouse fibroblasts, with small molecules. Based on alkaline phosphatase activity and other molecular markers, different compounds or their combinations are needed for converting the two different fibroblasts into DPC-LCs. Notably, we found that TTNPB alone can efficiently convert primary adult mouse fibroblasts into DPC-LCs. DPC-LCs generated from mouse fibroblasts showed a stronger hair-inducing capacity. Transcriptome analysis reveals that expression of genes associated with a hair-inducing capacity are increased in DPC-LCs. This pharmacological approach to generating functional dermal papilla cells may have many important implications for hair follicle regeneration and hair loss therapy.
Collapse
|
36
|
Abstract
Developing organs are shaped, in part, by physical interaction with their environment in the embryo. In recent years, technical advances in live-cell imaging and material science have greatly expanded our understanding of the mechanical forces driving organ formation. Here, we provide a broad overview of the types of forces generated during embryonic development and then focus on a subset of organs underlying our senses: the eyes, inner ears, nose and skin. The epithelia in these organs emerge from a common origin: the ectoderm germ layer; yet, they arrive at unique and complex forms over developmental time. We discuss exciting recent animal studies that show a crucial role for mechanical forces in, for example, the thickening of sensory placodes, the coiling of the cochlea and the lengthening of hair. Finally, we discuss how microfabricated organoid systems can now provide unprecedented insights into the physical principles of human development.
Collapse
Affiliation(s)
- Anh Phuong Le
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Plastic and Oral Surgery, Boston Children's Hospital, Boston, MA 02115, USA
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Jin Kim
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Plastic and Oral Surgery, Boston Children's Hospital, Boston, MA 02115, USA
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Karl R. Koehler
- Department of Otolaryngology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Plastic and Oral Surgery, Boston Children's Hospital, Boston, MA 02115, USA
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
37
|
Okamura DM, Nguyen ED, Beier DR, Majesky MW. Wound healing and regeneration in spiny mice (Acomys cahirinus). Curr Top Dev Biol 2022; 148:139-164. [DOI: 10.1016/bs.ctdb.2022.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
38
|
Frech S, Forsthuber A, Korosec A, Lipp K, Kozumov V, Lichtenberger BM. Hedgehog-signalling in papillary fibroblasts is essential for hair follicle regeneration during wound healing. J Invest Dermatol 2021; 142:1737-1748.e5. [PMID: 34922948 DOI: 10.1016/j.jid.2021.11.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/09/2021] [Accepted: 11/17/2021] [Indexed: 12/18/2022]
Abstract
Patients suffering from large scars such as burn victims not only encounter aesthetical challenges but also ongoing itching or pain that substantially deteriorates their quality of life. Skin appendages such as hair follicles (HFs) rarely regenerate within the healing wound. As they are crucial for skin homeostasis and the lack thereof constitutes one of the main limitations to scarless wound healing, their regeneration represents a major objective for regenerative medicine. Fibroblasts, the main resident cell type of the skin dermis, mediate embryonic HF morphogenesis and are particularly involved in wound healing as they orchestrate extracellular matrix (ECM) remodeling and collagen deposition in the wound bed. Importantly, dermal fibroblasts originate from two distinct developmental lineages with unique functions that differently mediate the response to epidermal signals such as Hedgehog (Hh)-signalling. In the present study, we show that Hh-signalling in the reticular fibroblast lineage promotes the initial phase of wound repair, possibly by modulating angiogenesis and fibroblast proliferation, while Hh-signalling in papillary fibroblasts is essential to induce de novo HF formation within the healing wound.
Collapse
Affiliation(s)
- Sophie Frech
- Skin & Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Lazarettgasse 14a, 1090 Vienna, Austria
| | - Agnes Forsthuber
- Skin & Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Lazarettgasse 14a, 1090 Vienna, Austria
| | - Ana Korosec
- Skin & Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Lazarettgasse 14a, 1090 Vienna, Austria
| | - Katharina Lipp
- Skin & Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Lazarettgasse 14a, 1090 Vienna, Austria
| | - Viktor Kozumov
- Skin & Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Lazarettgasse 14a, 1090 Vienna, Austria
| | - Beate M Lichtenberger
- Skin & Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Lazarettgasse 14a, 1090 Vienna, Austria.
| |
Collapse
|
39
|
Ankawa R, Fuchs Y. May the best wound WIHN: the hallmarks of wound-induced hair neogenesis. Curr Opin Genet Dev 2021; 72:53-60. [PMID: 34861514 DOI: 10.1016/j.gde.2021.10.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/05/2021] [Accepted: 10/21/2021] [Indexed: 01/06/2023]
Abstract
The hair follicle is a unique mini organ that undergoes continuous cycles of replenishment. While hair follicle formation was long thought to occur strictly during embryogenesis, it is now becoming increasingly clear that hair follicles can regenerate from the wound bed. Here, we provide an overview of the recent advancements in the field of Wound Induced Hair Neogenesis (WIHN) in mice. We briefly outline the hair follicle morphogenic process and discuss the major features of adult hair follicle regeneration. We examine the role of distinct cell types and review the contribution of specific signaling pathways to the WIHN phenotype. The phenomenon of neogenic hair regeneration provides an important platform, which may offer new insights into mammalian regeneration in the adult setting.
Collapse
Affiliation(s)
- Roi Ankawa
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Israel; Lorry Lokey Interdisciplinary Center for Life Sciences & Engineering, Technion Israel Institute of Technology, Israel; Technion Integrated Cancer Center, Technion Israel Institute of Technology, Haifa 3200, Israel
| | - Yaron Fuchs
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Israel; Lorry Lokey Interdisciplinary Center for Life Sciences & Engineering, Technion Israel Institute of Technology, Israel; Technion Integrated Cancer Center, Technion Israel Institute of Technology, Haifa 3200, Israel.
| |
Collapse
|
40
|
Peng H, Shindo K, Donahue RR, Gao E, Ahern BM, Levitan BM, Tripathi H, Powell D, Noor A, Elmore GA, Satin J, Seifert AW, Abdel-Latif A. Adult spiny mice (Acomys) exhibit endogenous cardiac recovery in response to myocardial infarction. NPJ Regen Med 2021; 6:74. [PMID: 34789749 PMCID: PMC8599698 DOI: 10.1038/s41536-021-00186-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 10/21/2021] [Indexed: 11/23/2022] Open
Abstract
Complex tissue regeneration is extremely rare among adult mammals. An exception, however, is the superior tissue healing of multiple organs in spiny mice (Acomys). While Acomys species exhibit the remarkable ability to heal complex tissue with minimal scarring, little is known about their cardiac structure and response to cardiac injury. In this study, we first examined baseline Acomys cardiac anatomy and function in comparison with commonly used inbred and outbred laboratory Mus strains (C57BL6 and CFW). While our results demonstrated comparable cardiac anatomy and function between Acomys and Mus, Acomys exhibited a higher percentage of cardiomyocytes displaying distinct characteristics. In response to myocardial infarction, all animals experienced a comparable level of initial cardiac damage. However, Acomys demonstrated superior ischemic tolerance and cytoprotection in response to injury as evidenced by cardiac functional stabilization, higher survival rate, and smaller scar size 50 days after injury compared to the inbred and outbred mouse strains. This phenomenon correlated with enhanced endothelial cell proliferation, increased angiogenesis, and medium vessel maturation in the peri-infarct and infarct regions. Overall, these findings demonstrate augmented myocardial preservation in spiny mice post-MI and establish Acomys as a new adult mammalian model for cardiac research.
Collapse
Affiliation(s)
- Hsuan Peng
- grid.266539.d0000 0004 1936 8438Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY USA
| | - Kazuhiro Shindo
- grid.266539.d0000 0004 1936 8438Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY USA
| | - Renée R. Donahue
- grid.266539.d0000 0004 1936 8438Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY USA
| | - Erhe Gao
- grid.264727.20000 0001 2248 3398The Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA USA
| | - Brooke M. Ahern
- grid.266539.d0000 0004 1936 8438Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY USA
| | - Bryana M. Levitan
- grid.266539.d0000 0004 1936 8438Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY USA ,grid.266539.d0000 0004 1936 8438Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY USA
| | - Himi Tripathi
- grid.266539.d0000 0004 1936 8438Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY USA
| | - David Powell
- grid.266539.d0000 0004 1936 8438Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY USA
| | - Ahmed Noor
- grid.266539.d0000 0004 1936 8438Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY USA
| | - Garrett A. Elmore
- grid.266539.d0000 0004 1936 8438Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY USA
| | - Jonathan Satin
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, USA.
| | - Ashley W. Seifert
- grid.266539.d0000 0004 1936 8438Department of Biology, University of Kentucky, Lexington, KY USA
| | - Ahmed Abdel-Latif
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY, USA. .,Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky, Lexington, KY, USA. .,The Lexington VA Medical Center, Lexington, KY, USA. .,Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
41
|
Abstract
Wounds in mammals commonly heal by scarring but retain latent capacity to regenerate. In this issue of Developmental Cell, Brewer, Nelson et al. reveal that heightened ability of spiny mice to regenerate after injury is attributed to molecular changes in the Hippo-YAP (yes-associated protein) pathway that protect wound fibroblasts from a persistent contractile state.
Collapse
Affiliation(s)
- Renzhi Hou
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Aliaksandr A Astrowski
- Institute of Biochemistry of Biologically Active Compounds of the National Academy of Sciences of Belarus, Grodna, Belarus
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
42
|
Brewer CM, Nelson BR, Wakenight P, Collins SJ, Okamura DM, Dong XR, Mahoney WM, McKenna A, Shendure J, Timms A, Millen KJ, Majesky MW. Adaptations in Hippo-Yap signaling and myofibroblast fate underlie scar-free ear appendage wound healing in spiny mice. Dev Cell 2021; 56:2722-2740.e6. [PMID: 34610329 DOI: 10.1016/j.devcel.2021.09.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 06/10/2021] [Accepted: 09/13/2021] [Indexed: 12/21/2022]
Abstract
Spiny mice (Acomys cahirinus) are terrestrial mammals that evolved unique scar-free regenerative wound-healing properties. Myofibroblasts (MFs) are the major scar-forming cell type in skin. We found that following traumatic injury to ear pinnae, MFs appeared rapidly in both Acomys and mouse yet persisted only in mouse. The timing of MF loss in Acomys correlated with wound closure, blastema differentiation, and nuclear localization of the Hippo pathway target protein Yap. Experiments in vitro revealed an accelerated PP2A-dependent dephosphorylation activity that maintained nuclear Yap in Acomys dermal fibroblasts (DFs) and was not detected in mouse or human DFs. Treatment of Acomys in vivo with the nuclear Yap-TEAD inhibitor verteporfin prolonged MF persistence and converted tissue regeneration to fibrosis. Forced Yap activity prevented and rescued TGF-β1-induced human MF formation in vitro. These results suggest that Acomys evolved modifications of Yap activity and MF fate important for scar-free regenerative wound healing in vivo.
Collapse
Affiliation(s)
- Chris M Brewer
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA; Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Branden R Nelson
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA.
| | - Paul Wakenight
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Sarah J Collins
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Daryl M Okamura
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Xiu Rong Dong
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - William M Mahoney
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Aaron McKenna
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, Seattle, WA 98195, USA
| | - Andrew Timms
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Kathleen J Millen
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA.
| | - Mark W Majesky
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA; Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101, USA.
| |
Collapse
|
43
|
Furnishing Wound Repair by the Subcutaneous Fascia. Int J Mol Sci 2021; 22:ijms22169006. [PMID: 34445709 PMCID: PMC8396603 DOI: 10.3390/ijms22169006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/10/2021] [Accepted: 08/17/2021] [Indexed: 12/28/2022] Open
Abstract
Mammals rapidly heal wounds through fibrous connective tissue build up and tissue contraction. Recent findings from mouse attribute wound healing to physical mobilization of a fibroelastic connective tissue layer that resides beneath the skin, termed subcutaneous fascia or superficial fascia, into sites of injury. Fascial mobilization assembles diverse cell types and matrix components needed for rapid wound repair. These observations suggest that the factors directly affecting fascial mobility are responsible for chronic skin wounds and excessive skin scarring. In this review, we discuss the link between the fascia's unique tissue anatomy, composition, biomechanical, and rheologic properties to its ability to mobilize its tissue assemblage. Fascia is thus at the forefront of tissue pathology and a better understanding of how it is mobilized may crystallize our view of wound healing alterations during aging, diabetes, and fibrous disease and create novel therapeutic strategies for wound repair.
Collapse
|