1
|
Mathisen AF, Larsen U, Kavli N, Unger L, Daian LM, Vacaru AM, Vacaru AM, Herrera PL, Ghila L, Chera S. Moderate beta-cell ablation triggers synergic compensatory mechanisms even in the absence of overt metabolic disruption. Commun Biol 2024; 7:833. [PMID: 38982170 PMCID: PMC11233560 DOI: 10.1038/s42003-024-06527-5] [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: 03/05/2024] [Accepted: 07/01/2024] [Indexed: 07/11/2024] Open
Abstract
Regeneration, the ability to replace injured tissues and organs, is a phenomenon commonly associated with lower vertebrates but is also observed in mammals, in specific tissues. In this study, we investigated the regenerative potential of pancreatic islets following moderate beta-cell loss in mice. Using a rapid model of moderate ablation, we observed a compensatory response characterized by transient inflammation and proliferation signatures, ultimately leading to the recovery of beta-cell identity and function. Interestingly, this proliferative response occurred independently of inflammation, as demonstrated in ablated immunodeficient mice. Furthermore, exposure to high-fat diet stimulated beta-cell proliferation but negatively impacted beta-cell function. In contrast, an equivalent slower ablation model revealed a delayed but similar proliferative response, suggesting proliferation as a common regenerative response. However, high-fat diet failed to promote proliferation in this model, indicating a differential response to metabolic stressors. Overall, our findings shed light on the complex interplay between beta-cell loss, inflammation, and stress in modulating pancreatic islet regeneration. Understanding these mechanisms could pave the way for novel therapeutic strategies based on beta-cell proliferation.
Collapse
Affiliation(s)
- Andreas Frøslev Mathisen
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ulrik Larsen
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Natalie Kavli
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Lucas Unger
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Laura Maria Daian
- BetaUpreg Research Group, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Andrei Mircea Vacaru
- BetaUpreg Research Group, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Ana-Maria Vacaru
- BetaUpreg Research Group, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Pedro Luis Herrera
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Luiza Ghila
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Simona Chera
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway.
| |
Collapse
|
2
|
Fang X, Zhou J, Yang Y, Li D, Wang B. Integrating scRNA-seq and bulk RNA-seq to explore the differentiation mechanism of human nail stem cells mediated by onychofibroblasts. Front Cell Dev Biol 2024; 12:1416780. [PMID: 38887517 PMCID: PMC11181305 DOI: 10.3389/fcell.2024.1416780] [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: 04/13/2024] [Accepted: 05/14/2024] [Indexed: 06/20/2024] Open
Abstract
Introduction: Nail stem cell (NSC) differentiation plays a vital role in maintaining nail homeostasis and facilitating digit regeneration. Recently, onychofibroblasts (OFs), specialized mesenchymal cells beneath the nail matrix, have emerged as potential regulators of NSC differentiation. However, limited understanding of OFs' cellular properties and transcriptomic profiles hinders our comprehension of their role. This study aims to characterize human OFs and investigate their involvement in NSC differentiation. Methods: Human OFs were isolated and characterized for their mesenchymal stem cell (MSC)-like phenotype through flow cytometry and multilineage differentiation assays. Bulk RNA-seq analysis was conducted on three samples of OFs and control fibroblasts from human nail units to delineate their molecular features. Integrated analysis with scRNA-seq data was performed to identify key signaling pathways involved in OF-induced NSC differentiation. Co-culture experiments, siRNA transfection, RT-qPCR, and immunocytochemistry were employed to investigate the effect of OF-derived soluble proteins on NSC differentiation. Drug treatments, RT-qPCR, western blotting, and immunocytochemistry were used to verify the regulation of candidate signaling pathways on NSC differentiation in vitro. Results: Human OFs exhibited slow cell cycle kinetics, expressed typical MSC markers, and demonstrated multilineage differentiation potential. Bulk RNA-seq analysis revealed differential gene expression in OFs compared to control fibroblasts, highlighting their role in coordinating nail development. Integrated analysis identified BMP4 as a pivotal signal for OFs to participate in NSC differentiation through mesenchymal-epithelial interactions, with the TGF-beta pathway possibly mediating this signal. OFs synthesized and secreted more BMP4 than control fibroblasts, and BMP4 derived from OFs induced NSC differentiation in a co-culture model. Recombinant human BMP4 activated the TGF-beta pathway in NSCs, leading to cell differentiation, while the BMP type I receptor inhibitor LDN193189 attenuated this effect. Discussion: This study characterizes the cellular and molecular features of human OFs, demonstrating their ability to regulate NSC differentiation via the TGF-beta signaling pathway. These findings establish a connection between the dermal microenvironment and NSC differentiation, suggesting the potential of OFs, in conjunction with NSCs, for developing novel therapies targeting nail and digit defects, even severe limb amputation.
Collapse
Affiliation(s)
- Xia Fang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Plastic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Jiateng Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Plastic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yating Yang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dawei Li
- Department of Plastic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Bin Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Inomata Y, Kawatani N, Yamashita H, Hattori F. Lgr6-expressing functional nail stem-like cells differentiated from human-induced pluripotent stem cells. PLoS One 2024; 19:e0303260. [PMID: 38743670 PMCID: PMC11093308 DOI: 10.1371/journal.pone.0303260] [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: 08/31/2023] [Accepted: 04/23/2024] [Indexed: 05/16/2024] Open
Abstract
The nail matrix containing stem cell populations produces nails and may contribute to fingertip regeneration. Nails are important tissues that maintain the functions of the hand and foot for handling objects and locomotion. Tumor chemotherapy impairs nail growth and, in many cases, loses them, although not permanently. In this report, we have achieved the successful differentiation of nail stem (NS)-like cells from human-induced pluripotent stem cells (iPSCs) via digit organoids by stepwise stimulation, tracing the molecular processes involved in limb development. Comprehensive mRNA sequencing analysis revealed that the digit organoid global gene expression profile fits human finger development. The NS-like cells expressed Lgr6 mRNA and protein and produced type-I keratin, KRT17, and type-II keratin, KRT81, which are abundant in nails. Furthermore, we succeeded in producing functional Lgr6-reporter human iPSCs. The reporter iPSC-derived Lgr6-positive cells also produced KRT17 and KRT81 proteins in the percutaneously transplanted region. To the best of our knowledge, this is the first report of NS-like cell differentiation from human iPSCs. Our differentiation method and reporter construct enable the discovery of drugs for nail repair and possibly fingertip-regenerative therapy.
Collapse
Affiliation(s)
- Yukino Inomata
- Innovative Regenerative Medicine, Graduate School of Medicine, Kansai Medical University, Hirakata city, Osaka, Japan
- Osaka College of High-Technology, Osaka City, Osaka, Japan
| | - Nano Kawatani
- Innovative Regenerative Medicine, Graduate School of Medicine, Kansai Medical University, Hirakata city, Osaka, Japan
- Osaka College of High-Technology, Osaka City, Osaka, Japan
| | - Hiromi Yamashita
- Innovative Regenerative Medicine, Graduate School of Medicine, Kansai Medical University, Hirakata city, Osaka, Japan
| | - Fumiyuki Hattori
- Innovative Regenerative Medicine, Graduate School of Medicine, Kansai Medical University, Hirakata city, Osaka, Japan
| |
Collapse
|
4
|
Tsissios G, Sallese A, Perez-Estrada JR, Tangeman JA, Chen W, Smucker B, Ratvasky SC, Grajales-Esquivel E, Martinez A, Visser KJ, Joven Araus A, Wang H, Simon A, Yun MH, Del Rio-Tsonis K. Macrophages modulate fibrosis during newt lens regeneration. Stem Cell Res Ther 2024; 15:141. [PMID: 38745238 PMCID: PMC11094960 DOI: 10.1186/s13287-024-03740-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 04/23/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Previous studies have suggested that macrophages are present during lens regeneration in newts, but their role in the process is yet to be elucidated. METHODS Here we generated a transgenic reporter line using the newt, Pleurodeles waltl, that traces macrophages during lens regeneration. Furthermore, we assessed early changes in gene expression during lens regeneration using two newt species, Notophthalmus viridescens and Pleurodeles waltl. Finally, we used clodronate liposomes to deplete macrophages during lens regeneration in both species and tested the effect of a subsequent secondary injury after macrophage recovery. RESULTS Macrophage depletion abrogated lens regeneration, induced the formation of scar-like tissue, led to inflammation, decreased iris pigment epithelial cell (iPEC) proliferation, and increased rates of apoptosis in the eye. Some of these phenotypes persisted throughout the last observation period of 100 days and could be attenuated by exogenous FGF2 administration. A distinct transcript profile encoding acute inflammatory effectors was established for the dorsal iris. Reinjury of the newt eye alleviated the effects of macrophage depletion, including the resolution of scar-like tissue, and re-initiated the regeneration process. CONCLUSIONS Together, our findings highlight the importance of macrophages for facilitating a pro-regenerative environment in the newt eye by regulating fibrotic responses, modulating the overall inflammatory landscape, and maintaining the proper balance of early proliferation and late apoptosis of the iPECs.
Collapse
Affiliation(s)
- Georgios Tsissios
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
| | - Anthony Sallese
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
| | - J Raul Perez-Estrada
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
| | - Jared A Tangeman
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
| | - Weihao Chen
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, OH, USA
| | - Byran Smucker
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
- Department of Statistics, Miami University, Oxford, OH, USA
| | - Sophia C Ratvasky
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
| | - Erika Grajales-Esquivel
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
| | - Arielle Martinez
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
| | - Kimberly J Visser
- CRTD/ Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Alberto Joven Araus
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Hui Wang
- Center for Visual Sciences at, Miami University, Oxford, OH, USA
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, OH, USA
| | - András Simon
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Maximina H Yun
- CRTD/ Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden, Germany
| | - Katia Del Rio-Tsonis
- Department of Biology, Miami University, Oxford, OH, USA.
- Center for Visual Sciences at, Miami University, Oxford, OH, USA.
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA.
| |
Collapse
|
5
|
Tan FH, Bronner ME. Regenerative loss in the animal kingdom as viewed from the mouse digit tip and heart. Dev Biol 2024; 507:44-63. [PMID: 38145727 PMCID: PMC10922877 DOI: 10.1016/j.ydbio.2023.12.008] [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: 08/06/2023] [Revised: 11/30/2023] [Accepted: 12/19/2023] [Indexed: 12/27/2023]
Abstract
The myriad regenerative abilities across the animal kingdom have fascinated us for centuries. Recent advances in developmental, molecular, and cellular biology have allowed us to unearth a surprising diversity of mechanisms through which these processes occur. Developing an all-encompassing theory of animal regeneration has thus proved a complex endeavor. In this chapter, we frame the evolution and loss of animal regeneration within the broad developmental constraints that may physiologically inhibit regenerative ability across animal phylogeny. We then examine the mouse as a model of regeneration loss, specifically the experimental systems of the digit tip and heart. We discuss the digit tip and heart as a positionally-limited system of regeneration and a temporally-limited system of regeneration, respectively. We delve into the physiological processes involved in both forms of regeneration, and how each phase of the healing and regenerative process may be affected by various molecular signals, systemic changes, or microenvironmental cues. Lastly, we also discuss the various approaches and interventions used to induce or improve the regenerative response in both contexts, and the implications they have for our understanding regenerative ability more broadly.
Collapse
Affiliation(s)
- Fayth Hui Tan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
6
|
Tsissios G, Sallese A, Perez-Estrada JR, Tangeman JA, Chen W, Smucker B, Ratvasky SC, Grajales-Esquive EL, Martinez A, Visser KJ, Araus AJ, Wang H, Simon A, Yun MH, Rio-Tsonis KD. Macrophages modulate fibrosis during newt lens regeneration. RESEARCH SQUARE 2023:rs.3.rs-3603645. [PMID: 38045376 PMCID: PMC10690311 DOI: 10.21203/rs.3.rs-3603645/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Background Previous studies indicated that macrophages play a role during lens regeneration in newts, but their function has not been tested experimentally. Methods Here we generated a transgenic newt reporter line in which macrophages can be visualized in vivo. Using this new tool, we analyzed the location of macrophages during lens regeneration. We uncovered early gene expression changes using bulk RNAseq in two newt species, Notophthalmus viridescens and Pleurodeles waltl. Next, we used clodronate liposomes to deplete macrophages, which inhibited lens regeneration in both newt species. Results Macrophage depletion induced the formation of scar-like tissue, an increased and sustained inflammatory response, an early decrease in iris pigment epithelial cell (iPEC) proliferation and a late increase in apoptosis. Some of these phenotypes persisted for at least 100 days and could be rescued by exogenous FGF2. Re-injury alleviated the effects of macrophage depletion and re-started the regeneration process. Conclusions Together, our findings highlight the importance of macrophages in facilitating a pro-regenerative environment in the newt eye, helping to resolve fibrosis, modulating the overall inflammatory landscape and maintaining the proper balance of early proliferation and late apoptosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Maximina H Yun
- Dresden University of Technology: Technische Universitat Dresden
| | | |
Collapse
|
7
|
Lin YL, Yu L, Yan M, Zimmel K, Qureshi O, Imholt F, Li T, Ivanov I, Brunauer R, Dawson L, Muneoka K. Induced regeneration of articular cartilage - identification of a dormant regeneration program for a non-regenerative tissue. Development 2023; 150:dev201894. [PMID: 37882667 PMCID: PMC10651102 DOI: 10.1242/dev.201894] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 10/09/2023] [Indexed: 10/27/2023]
Abstract
A mouse organoid culture model was developed to regenerate articular cartilage by sequential treatment with BMP2 and BMP9 (or GDF2) that parallels induced joint regeneration at digit amputation wounds in vivo. BMP9-induced chondrogenesis was used to identify clonal cell lines for articular chondrocyte and hypertrophic chondrocyte progenitor cells from digit fibroblasts. A protocol that includes cell aggregation enhanced by BMP2 followed by BMP9-induced chondrogenesis resulted in the differentiation of organized layers of articular chondrocytes, similar to the organization of middle and deep zones of articular cartilage in situ, and retained a differentiated phenotype following transplantation. In addition, the differentiation of a non-chondrogenic connective tissue layer containing articular chondrocyte progenitor cells demonstrated that progenitor cell sequestration is coupled with articular cartilage differentiation at a clonal level. The studies identify a dormant endogenous regenerative program for a non-regenerative tissue in which fibroblast-derived progenitor cells can be induced to initiate morphogenetic and differentiative programs that include progenitor cell sequestration. The identification of dormant regenerative programs in non-regenerative tissues such as articular cartilage represents a novel strategy that integrates regeneration biology with regenerative medicine.
Collapse
Affiliation(s)
- Yu-Lieh Lin
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Ling Yu
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Mingquan Yan
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Katherine Zimmel
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Osama Qureshi
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Felisha Imholt
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Tao Li
- Department of Hand Surgery, Union Hospital, Tongli Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, People's Republic of China
| | - Ivan Ivanov
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Regina Brunauer
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Lindsay Dawson
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Ken Muneoka
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
8
|
Li C. Deer antler renewal gives insights into mammalian epimorphic regeneration. CELL REGENERATION (LONDON, ENGLAND) 2023; 12:26. [PMID: 37490254 PMCID: PMC10368610 DOI: 10.1186/s13619-023-00169-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 07/02/2023] [Indexed: 07/26/2023]
Abstract
Deer antlers are the only known mammalian organ that, once lost, can fully grow back naturally. Hence, the antler offers a unique opportunity to learn how nature has solved the problem of mammalian epimorphic regeneration (EpR). Comprehensive comparisons amongst different types of EpR reveal that antler renewal is fundamentally different from that in lower vertebrates such as regeneration of the newt limb. Surprisingly, antler renewal is comparable to wound healing over a stump of regeneration-incompetent digit/limb, bone fracture repair, and to a lesser extent to digit tip regeneration in mammals. Common to all these mammalian cases of reaction to the amputation/mechanical trauma is the response of the periosteal cells at the distal end/injury site with formation of a circumferential cartilaginous callus (CCC). Interestingly, whether the CCC can proceed to the next stage to transform to a blastema fully depends on the presence of an interactive partner. The actual form of the partner can vary in different cases with the nail organ in digit tip EpR, the opposing callus in bone fracture repair, and the closely associated enveloping skin in antler regeneration. Due to absence of such an interactive partner, the CCC of a mouse/rat digit/limb stump becomes involuted gradually. Based on these discoveries, we created an interactive partner for the rat digit/limb stump through surgically removal of the interposing layers of loose connective tissue and muscle between the resultant CCC and the enveloping skin after amputation and by forcefully bonding two tissue types tightly together. In so doing partial regeneration of the limb stump occurred. In summary, if EpR in humans is to be realized, then I envisage that it would be more likely in a manner akin to antler regeneration rather to that of lower vertebrates such as newt limbs.
Collapse
Affiliation(s)
- Chunyi Li
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, 130600, China.
- Jilin Provincial Key Laboratory of Deer Antler Biology, Changchun, 130600, China.
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, 130000, China.
| |
Collapse
|
9
|
Tsissios G, Sallese A, Perez-Estrada JR, Tangeman JA, Chen W, Smucker B, Ratvasky SC, Grajales-Esquivel E, Martinez A, Visser KJ, Araus AJ, Wang H, Simon A, Yun MH, Rio-Tsonis KD. Macrophages modulate fibrosis during newt lens regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.04.543633. [PMID: 37333184 PMCID: PMC10274724 DOI: 10.1101/2023.06.04.543633] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Previous studies indicated that macrophages play a role during lens regeneration in newts, but their function has not been tested experimentally. Here we generated a transgenic newt reporter line in which macrophages can be visualized in vivo. Using this new tool, we analyzed the location of macrophages during lens regeneration. We uncovered early gene expression changes using bulk RNAseq in two newt species, Notophthalmus viridescens and Pleurodeles waltl. Next, we used clodronate liposomes to deplete macrophages, which inhibited lens regeneration in both newt species. Macrophage depletion induced the formation of scar-like tissue, an increased and sustained inflammatory response, an early decrease in iris pigment epithelial cell (iPEC) proliferation and a late increase in apoptosis. Some of these phenotypes persisted for at least 100 days and could be rescued by exogenous FGF2. Re-injury alleviated the effects of macrophage depletion and re-started the regeneration process. Together, our findings highlight the importance of macrophages in facilitating a pro-regenerative environment in the newt eye, helping to resolve fibrosis, modulating the overall inflammatory landscape and maintaining the proper balance of early proliferation and late apoptosis.
Collapse
Affiliation(s)
- Georgios Tsissios
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
| | - Anthony Sallese
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at Miami University, Oxford, OH, USA
| | - J Raul Perez-Estrada
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at Miami University, Oxford, OH, USA
| | - Jared A Tangeman
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
| | - Weihao Chen
- Center for Visual Sciences at Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, OH, USA
| | - Byran Smucker
- Center for Visual Sciences at Miami University, Oxford, OH, USA
- Department of Statistics, Miami University, Oxford, OH, USA
| | - Sophia C Ratvasky
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
| | - Erika Grajales-Esquivel
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at Miami University, Oxford, OH, USA
| | - Arielle Martinez
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at Miami University, Oxford, OH, USA
| | - Kimberly J Visser
- CRTD Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Alberto Joven Araus
- Karolinska Institute, Department of Cell and Molecular Biology, Stockholm, Sweden
| | - Hui Wang
- Center for Visual Sciences at Miami University, Oxford, OH, USA
- Department of Chemical, Paper and Biomedical Engineering, Miami University, Oxford, OH, USA
| | - Andras Simon
- Karolinska Institute, Department of Cell and Molecular Biology, Stockholm, Sweden
| | - Maximina H Yun
- CRTD Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden, Germany
| | - Katia Del Rio-Tsonis
- Department of Biology, Miami University, Oxford, OH, USA
- Center for Visual Sciences at Miami University, Oxford, OH, USA
- Cellular Molecular and Structural Biology Program, Miami University, Oxford, OH, USA
| |
Collapse
|
10
|
Jou V, Lehoczky JA. Toeing the line between regeneration and fibrosis. Front Cell Dev Biol 2023; 11:1217185. [PMID: 37325560 PMCID: PMC10267333 DOI: 10.3389/fcell.2023.1217185] [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: 05/05/2023] [Accepted: 05/23/2023] [Indexed: 06/17/2023] Open
Abstract
Understanding the remarkable capacity of vertebrates to naturally regenerate injured body parts has great importance for potential translation into human therapeutic applications. As compared to other vertebrates, mammals have low regenerative capacity for composite tissues like the limb. However, some primates and rodents can regenerate the distal tips of their digits following amputation, indicating that at least very distal mammalian limb tissues are competent for innate regeneration. It follows that successful digit tip regenerative outcome is highly dependent on the location of the amputation; those proximal to the position of the nail organ do not regenerate and result in fibrosis. This distal regeneration versus proximal fibrosis duality of the mouse digit tip serves as a powerful model to investigate the driving factors in determining each process. In this review, we present the current understanding of distal digit tip regeneration in the context of cellular heterogeneity and the potential for different cell types to function as progenitor cells, in pro-regenerative signaling, or in moderating fibrosis. We then go on to discuss these themes in the context of what is known about proximal digit fibrosis, towards generating hypotheses for these distinct healing processes in the distal and proximal mouse digit.
Collapse
Affiliation(s)
- Vivian Jou
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, United States
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Boston, MA, United States
| | - Jessica A. Lehoczky
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Boston, MA, United States
| |
Collapse
|
11
|
Fuiten AM, Yoshimoto Y, Shukunami C, Stadler HS. Digits in a dish: An in vitro system to assess the molecular genetics of hand/foot development at single-cell resolution. Front Cell Dev Biol 2023; 11:1135025. [PMID: 36994104 PMCID: PMC10040768 DOI: 10.3389/fcell.2023.1135025] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 02/21/2023] [Indexed: 03/16/2023] Open
Abstract
In vitro models allow for the study of developmental processes outside of the embryo. To gain access to the cells mediating digit and joint development, we identified a unique property of undifferentiated mesenchyme isolated from the distal early autopod to autonomously re-assemble forming multiple autopod structures including: digits, interdigital tissues, joints, muscles and tendons. Single-cell transcriptomic analysis of these developing structures revealed distinct cell clusters that express canonical markers of distal limb development including: Col2a1, Col10a1, and Sp7 (phalanx formation), Thbs2 and Col1a1 (perichondrium), Gdf5, Wnt5a, and Jun (joint interzone), Aldh1a2 and Msx1 (interdigital tissues), Myod1 (muscle progenitors), Prg4 (articular perichondrium/articular cartilage), and Scx and Tnmd (tenocytes/tendons). Analysis of the gene expression patterns for these signature genes indicates that developmental timing and tissue-specific localization were also recapitulated in a manner similar to the initiation and maturation of the developing murine autopod. Finally, the in vitro digit system also recapitulates congenital malformations associated with genetic mutations as in vitro cultures of Hoxa13 mutant mesenchyme produced defects present in Hoxa13 mutant autopods including digit fusions, reduced phalangeal segment numbers, and poor mesenchymal condensation. These findings demonstrate the robustness of the in vitro digit system to recapitulate digit and joint development. As an in vitro model of murine digit and joint development, this innovative system will provide access to the developing limb tissues facilitating studies to discern how digit and articular joint formation is initiated and how undifferentiated mesenchyme is patterned to establish individual digit morphologies. The in vitro digit system also provides a platform to rapidly evaluate treatments aimed at stimulating the repair or regeneration of mammalian digits impacted by congenital malformation, injury, or disease.
Collapse
Affiliation(s)
- Allison M. Fuiten
- Research Center, Shriners Children’s, Portland, OR, United States
- Department of Orthopaedics and Rehabilitation, Oregon Health and Science University, Portland, OR, United States
| | - Yuki Yoshimoto
- Department of Molecular Biology and Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Chisa Shukunami
- Department of Molecular Biology and Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - H. Scott Stadler
- Research Center, Shriners Children’s, Portland, OR, United States
- Department of Orthopaedics and Rehabilitation, Oregon Health and Science University, Portland, OR, United States
- *Correspondence: H. Scott Stadler,
| |
Collapse
|
12
|
Abarca‐Buis RF, Krötzsch E. Proximal ear hole injury heals by limited regeneration during the early postnatal phase in mice. J Anat 2023; 242:402-416. [PMID: 36317926 PMCID: PMC9919478 DOI: 10.1111/joa.13782] [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: 04/08/2022] [Revised: 10/06/2022] [Accepted: 10/06/2022] [Indexed: 02/12/2023] Open
Abstract
Ear pinna is a particular feature of mammals that shows several repair responses depending on age. Two millimeter hole made in the pinna of middle-aged female mice heals due to partial reconstitution of new tissues (limited regeneration), whereas a hole punched in the ear of young mice forms a scar tissue. In these studies, the injury is made in the center of the ear pinna, but little is known about the type of reparative response along the proximodistal polarity of the ear. This study evaluated the effect of pinna polarity, age, and sex in the ear hole-repairing response in Balb/c mice. Proximal injuries were repaired more efficiently by limited regeneration than wounds made in the middle region. Non-injured ear histological analysis revealed a higher presence of muscle, adipose tissue, cartilage, and larger blood vessels in the proximal ear area, which could influence ear hole closure by limited regeneration. To evaluate the healing response during ear growth, we punched a standard hole in the proximal area of the ear on postnatal day 21 and 8-month-old mice (adults). Thirty-five days after the wound, both groups reached the same wound closure, despite the greater proportional size of holes made in the younger mice. Ear growth also improved ear hole closure in male mice. These results suggest that ear growth accelerates hole closure, providing an example of enhanced regenerative abilities in growing structures. Finally, hole closure kinetics in the growing ear indicated an early re-differentiation phase exhibited at 14 days post-wound. In conclusion, ear topography and growth positively influenced the healing response to ear holes, making it a tractable model to study in mammals.
Collapse
Affiliation(s)
- René Fernando Abarca‐Buis
- Laboratory of Connective Tissue, Centro Nacional de Investigación y Atención de QuemadosInstituto Nacional de Rehabilitación “Luís Guillermo Ibarra Ibarra”Ciudad de MéxicoMexico
| | - Edgar Krötzsch
- Laboratory of Connective Tissue, Centro Nacional de Investigación y Atención de QuemadosInstituto Nacional de Rehabilitación “Luís Guillermo Ibarra Ibarra”Ciudad de MéxicoMexico
| |
Collapse
|
13
|
Castilla-Ibeas A, Zdral S, Galán L, Haro E, Allou L, Campa VM, Icardo JM, Mundlos S, Oberg KC, Ros MA. Failure of digit tip regeneration in the absence of Lmx1b suggests Lmx1b functions disparate from dorsoventral polarity. Cell Rep 2023; 42:111975. [PMID: 36641754 DOI: 10.1016/j.celrep.2022.111975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 11/07/2022] [Accepted: 12/22/2022] [Indexed: 01/15/2023] Open
Abstract
Mammalian digit tip regeneration is linked to the presence of nail tissue, but a nail-explicit model is missing. Here, we report that nail-less double-ventral digits of ΔLARM1/2 mutants that lack limb-specific Lmx1b enhancers fail to regenerate. To separate the nail's effect from the lack of dorsoventral (DV) polarity, we also interrogate double-dorsal double-nail digits and show that they regenerate. Thus, DV polarity is not a prerequisite for regeneration, and the nail requirement is supported. Transcriptomic comparison between wild-type and non-regenerative ΔLARM1/2 mutant blastemas reveals differential upregulation of vascularization and connective tissue functional signatures in wild type versus upregulation of inflammation in the mutant. These results, together with the finding of Lmx1b expression in the postnatal dorsal dermis underneath the nail and uniformly in the regenerative blastema, open the possibility of additional Lmx1b roles in digit tip regeneration, in addition to the indirect effect of mediating the formation of the nail.
Collapse
Affiliation(s)
- Alejandro Castilla-Ibeas
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC; CSIC-SODERCAN-UC), Santander, Spain
| | - Sofía Zdral
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC; CSIC-SODERCAN-UC), Santander, Spain
| | - Laura Galán
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC; CSIC-SODERCAN-UC), Santander, Spain
| | - Endika Haro
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC; CSIC-SODERCAN-UC), Santander, Spain
| | - Lila Allou
- RG Development & Disease, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Víctor M Campa
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC; CSIC-SODERCAN-UC), Santander, Spain
| | - Jose M Icardo
- Departamento de Anatomía y Biología Celular, Universidad de Cantabria, Santander, Spain
| | - Stefan Mundlos
- RG Development & Disease, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Kerby C Oberg
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Marian A Ros
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC; CSIC-SODERCAN-UC), Santander, Spain.
| |
Collapse
|
14
|
Jaramillo J, Taylor C, McCarley R, Berger M, Busse E, Sammarco MC. Oxaloacetate enhances and accelerates regeneration in young mice by promoting proliferation and mineralization. Front Cell Dev Biol 2023; 11:1117836. [PMID: 36910154 PMCID: PMC9999028 DOI: 10.3389/fcell.2023.1117836] [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: 12/06/2022] [Accepted: 02/08/2023] [Indexed: 03/14/2023] Open
Abstract
Cell metabolism coordinates the biochemical reactions that produce carbon and ATP in order for the cell to proliferate, differentiate, and respond to environmental changes. Cell type determines metabolic demand, so proliferating skeletal progenitors and differentiated osteoblasts exhibit different levels of cell metabolism. Limb regeneration is an energetically demanding process that involves multiple types of tissues and cell functions over time. Dysregulation of cell metabolism in aged mice results in impaired regeneration, a defect that can be rescued in part by the administration of oxaloacetate (OAA). A better understanding of how cell metabolism regulates regeneration in general, and how these changes can be modulated to benefit potential regenerative strategies in the future is needed. Here we sought to better understand the effects of OAA on young mice and determine whether the same mechanism could be tapped to improve regeneration without an aged-defect. We also asked which dosing time periods were most impactful for promoting regenerative outcomes, and whether these effects were sustained after dosing was stopped. Consistent with our findings in aged mice we found that OAA enhanced regeneration by accelerating bone growth, even beyond control measures, by increasing trabecular thickness, decreasing trabecular spacing, and improving the patterning by decreasing the taper, making the regenerated bone more like an unamputated digit. Our data suggests that the decrease in spacing, an improvement over aged mice, may be due to a decrease in hypoxia-driven vasculature. Our findings suggest that OAA, and similar metabolites, may be a strong tool to promote regenerative strategies and investigate the mechanisms that link cell metabolism and regeneration.
Collapse
Affiliation(s)
- Josue Jaramillo
- Department of Surgery, Tulane School of Medicine, New Orleans, LA, United States
| | - Caroline Taylor
- Department of Surgery, Tulane School of Medicine, New Orleans, LA, United States
| | - Rachel McCarley
- Department of Surgery, Tulane School of Medicine, New Orleans, LA, United States
| | - Melissa Berger
- Department of Surgery, Tulane School of Medicine, New Orleans, LA, United States
| | - Emily Busse
- Department of Surgery, Tulane School of Medicine, New Orleans, LA, United States
| | - Mimi C Sammarco
- Department of Surgery, Tulane School of Medicine, New Orleans, LA, United States
| |
Collapse
|
15
|
Ketcham PD, Imholt F, Yan M, Smith HM, Asrar S, Yu L, Dolan CP, Qureshi O, Lin YL, Xia I, Hall PC, Falck AR, Sherman KM, Gaddy D, Suva LJ, Muneoka K, Brunauer R, Dawson LA. Microcomputed tomography staging of bone histolysis in the regenerating mouse digit. Wound Repair Regen 2023; 31:17-27. [PMID: 36177656 DOI: 10.1111/wrr.13054] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/10/2022] [Accepted: 09/14/2022] [Indexed: 01/25/2023]
Abstract
Humans and mice have the ability to regenerate the distal digit tip, the terminal phalanx (P3) in response to amputation. What distinguishes P3 regeneration from regenerative failure is formation of the blastema, a proliferative structure that undergoes morphogenesis to regenerate the amputated tissues. P3 regeneration is characterised by the phases of inflammation, tissue histolysis and expansive bone degradation with simultaneous blastema formation, wound closure and finally blastemal differentiation to restore the amputated structures. While each regenerating digit faithfully progresses through all phases of regeneration, phase progression has traditionally been delineated by time, that is, days postamputation (DPA), yet there is widespread variability in the timing of the individual phases. To diminish variability between digits during tissue histolysis and blastema formation, we have established an in-vivo method using microcomputed tomography (micro CT) scanning to identify five distinct stages of the early regeneration response based on anatomical changes of the digit stump. We report that categorising the initial phases of digit regeneration by stage rather than time greatly diminishes the variability between digits with respect to changes in bone volume and length. Also, stages correlate with the levels of cell proliferation, osteoclast recruitment and osteoprogenitor cell recruitment. Importantly, micro CT staging provides a means to estimate open versus closed digit wounds. We demonstrate two spatially distinct and stage specific bone repair/regeneration responses that occur during P3 regeneration. Collectively, these studies showcase the utility of micro CT imaging to infer the composition of radiolucent soft tissues during P3 blastema formation. Specifically, the staging system identifies the onset of cell proliferation, osteoclastogenesis, osteoprogenitor recruitment, the spatial initiation of de novo bone formation and epidermal closure.
Collapse
Affiliation(s)
- Paulina D Ketcham
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Felisha Imholt
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA.,Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon, USA
| | - Mingquan Yan
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Hannah M Smith
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Shabistan Asrar
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Ling Yu
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Connor P Dolan
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA.,DoD-VA Extremity Trauma and Amputation Centre of Excellence, Bethesda, Maryland, USA.,Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Centre, Bethesda, Maryland, USA
| | - Osama Qureshi
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Yu-Lieh Lin
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Ian Xia
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Patrick C Hall
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Alyssa R Falck
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Kirby M Sherman
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Dana Gaddy
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Larry J Suva
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Ken Muneoka
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Regina Brunauer
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Lindsay A Dawson
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
16
|
Mahmud N, Eisner C, Purushothaman S, Storer MA, Kaplan DR, Miller FD. Nail-associated mesenchymal cells contribute to and are essential for dorsal digit tip regeneration. Cell Rep 2022; 41:111853. [PMID: 36543145 DOI: 10.1016/j.celrep.2022.111853] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 10/05/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
Here, we ask why the nail base is essential for mammalian digit tip regeneration, focusing on the inductive nail mesenchyme. We identify a transcriptional signature for these cells that includes Lmx1b and show that the Lmx1b-expressing nail mesenchyme is essential for blastema formation. We use a combination of Lmx1bCreERT2-based lineage-tracing and single-cell transcriptional analyses to show that the nail mesenchyme contributes cells for two pro-regenerative mechanisms. One group of cells maintains their identity and regenerates the new nail mesenchyme. A second group contributes specifically to the dorsal blastema, loses their nail mesenchyme phenotype, acquires a blastema transcriptional state that is highly similar to blastema cells of other origins, and ultimately contributes to regeneration of the dorsal but not ventral dermis and bone. Thus, the regenerative necessity for an intact nail base is explained, at least in part, by a requirement for the inductive nail mesenchyme.
Collapse
Affiliation(s)
- Neemat Mahmud
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, ON M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5G 1A8, Canada
| | - Christine Eisner
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, ON M5G 1L7, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Sruthi Purushothaman
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, ON M5G 1L7, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Mekayla A Storer
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, ON M5G 1L7, Canada
| | - David R Kaplan
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1A8, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z, Canada
| | - Freda D Miller
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, ON M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5G 1A8, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1A8, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z, Canada.
| |
Collapse
|
17
|
Hoffseth KF, Busse E, Lacey M, Sammarco MC. Evaluating differences in Young's Modulus of regenerated and uninjured mouse digit bone through microCT density-based calculation and nanoindentation testing. J Biomech 2022; 143:111271. [PMID: 36095912 PMCID: PMC9947921 DOI: 10.1016/j.jbiomech.2022.111271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 07/12/2022] [Accepted: 08/21/2022] [Indexed: 02/05/2023]
Abstract
The mouse digit tip amputation model is an excellent model of bone regeneration, but its size and shape present an obstacle for biomechanical testing. As a result, assessing the structural quality of the regenerated bone in this model has focused on mineral density and bone architecture analysis. Here we describe an image-processing based method for assessment of mechanical properties in the regenerated digit by using micro-computed tomography mineral density data to calculate spatially discrete Young's modulus values throughout the entire distal third phalange. Further, we validate this method through comparison to nanoindentation-measured values for Young's modulus. Application to a set of regenerated and unamputated digits shows that regenerated bone has a lower Young's modulus compared to the uninjured digit, with a similar trend for experimental hardness values. Importantly, this method heightens the utility of the digit regeneration model, allows for more impactful treatment evaluation using the model, and introduces an analysis platform that can be used for other bones that do not conform to a standard long-bone model.
Collapse
Affiliation(s)
- Kevin F. Hoffseth
- Department of Biological & Agricultural Engineering, Louisiana State University, 149 E.B. Doran Building, Baton Rouge, LA 70803, USA,Corresponding authors. (K.F. Hoffseth), (M.C. Sammarco)
| | - Emily Busse
- Department of Surgery, Tulane School of Medicine, 1430 Tulane Ave, New Orleans, LA 70112, USA
| | - Michelle Lacey
- Department of Mathematics, Tulane University, 6823 St. Charles Avenue, New Orleans, LA 70118, USA
| | - Mimi C. Sammarco
- Department of Surgery, Tulane School of Medicine, 1430 Tulane Ave, New Orleans, LA 70112, USA,Corresponding authors. (K.F. Hoffseth), (M.C. Sammarco)
| |
Collapse
|
18
|
Tower RJ, Bancroft AC, Chowdary AR, Barnes S, Edwards NJ, Pagani CA, Dawson LA, Levi B. Single-cell mapping of regenerative and fibrotic healing responses after musculoskeletal injury. Stem Cell Reports 2022; 17:2334-2348. [PMID: 36150381 PMCID: PMC9561541 DOI: 10.1016/j.stemcr.2022.08.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/25/2022] [Accepted: 08/25/2022] [Indexed: 11/09/2022] Open
Abstract
After injury, a cascade of events repairs the damaged tissue, including expansion and differentiation of the progenitor pool and redeposition of matrix. To guide future wound regeneration strategies, we compared single-cell sequencing of regenerative (third phalangeal element [P3]) and fibrotic (second phalangeal element [P2]) digit tip amputation (DTA) models as well as traumatic heterotopic ossification (HO; aberrant). Analyses point to a common initial response to injury, including expansion of progenitors, redeposition of matrix, and activation of transforming growth factor β (TGF-β) and WNT pathways. Surprisingly, fibrotic P2 DTA showed greater transcriptional similarity to HO than to regenerative P3 DTA, suggesting that gene expression more strongly correlates with healing outcome than with injury type or cell origin. Differential analysis and immunostaining revealed altered activation of inflammatory pathways, such as the complement pathway, in the progenitor cells. These data suggests that common pathways are activated in response to damage but are fine tuned within each injury. Modulating these pathways may shift the balance toward regenerative outcomes. Regenerative and fibrotic injuries share common early response mechanisms Transcriptomes correlate with healing outcome more than injury type or cell source Matrix composition after injury-induced tissue repair is highly injury type dependent Inflammatory cascades are activated in immune and mesenchymal cells
Collapse
Affiliation(s)
- Robert J Tower
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Alec C Bancroft
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ashish R Chowdary
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Spencer Barnes
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Bioinformatics Core, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Nicole J Edwards
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Chase A Pagani
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lindsay A Dawson
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Benjamin Levi
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
19
|
Vinestock RC, Felsenthal N, Assaraf E, Katz E, Rubin S, Heinemann-Yerushalmi L, Krief S, Dezorella N, Levin-Zaidman S, Tsoory M, Thomopoulos S, Zelzer E. Neonatal Enthesis Healing Involves Noninflammatory Acellular Scar Formation through Extracellular Matrix Secretion by Resident Cells. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1122-1135. [PMID: 35659946 PMCID: PMC9379688 DOI: 10.1016/j.ajpath.2022.05.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/19/2022] [Accepted: 05/04/2022] [Indexed: 06/15/2023]
Abstract
Wound healing typically recruits the immune and vascular systems to restore tissue structure and function. However, injuries to the enthesis, a hypocellular and avascular tissue, often result in fibrotic scar formation and loss of mechanical properties, severely affecting musculoskeletal function and life quality. This raises questions about the healing capabilities of the enthesis. Herein, this study established an injury model to the Achilles entheses of neonatal mice to study the effectiveness of early-age enthesis healing. Histology and immunohistochemistry analyses revealed an atypical process that did not involve inflammation or angiogenesis. Instead, healing was mediated by secretion of collagen types I and II by resident cells, which formed a permanent hypocellular and avascular scar. Transmission electron microscopy showed that the cellular response to injury, including endoplasmic reticulum stress, autophagy, and cell death, varied between the tendon and cartilage ends of the enthesis. Single-molecule in situ hybridization, immunostaining, and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assays verified these differences. Finally, gait analysis showed that these processes effectively restored function of the injured leg. These findings reveal a novel healing mechanism in neonatal entheses, whereby local extracellular matrix secretion by resident cells forms an acellular extracellular matrix deposit without inflammation, allowing gait restoration. These insights into the healing mechanism of a complex transitional tissue may lead to new therapeutic strategies for adult enthesis injuries.
Collapse
Affiliation(s)
- Ron C Vinestock
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Neta Felsenthal
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Assaraf
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Eldad Katz
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Sarah Rubin
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | | | - Sharon Krief
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Nili Dezorella
- Department of Electron Microscopy Unit, Weizmann Institute of Science, Rehovot, Israel
| | - Smadar Levin-Zaidman
- Department of Electron Microscopy Unit, Weizmann Institute of Science, Rehovot, Israel
| | - Michael Tsoory
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Stavros Thomopoulos
- Department of Orthopedic Surgery, Columbia University, New York, New York; Department of Biomedical Engineering, Columbia University, New York, New York
| | - Elazar Zelzer
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
20
|
Tower RJ, Busse E, Jaramillo J, Lacey M, Hoffseth K, Guntur AR, Simkin J, Sammarco MC. Spatial transcriptomics reveals metabolic changes underly age-dependent declines in digit regeneration. eLife 2022; 11:71542. [PMID: 35616636 PMCID: PMC9135401 DOI: 10.7554/elife.71542] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 05/18/2022] [Indexed: 12/12/2022] Open
Abstract
De novo limb regeneration after amputation is restricted in mammals to the distal digit tip. Central to this regenerative process is the blastema, a heterogeneous population of lineage-restricted, dedifferentiated cells that ultimately orchestrates regeneration of the amputated bone and surrounding soft tissue. To investigate skeletal regeneration, we made use of spatial transcriptomics to characterize the transcriptional profile specifically within the blastema. Using this technique, we generated a gene signature with high specificity for the blastema in both our spatial data, as well as other previously published single-cell RNA-sequencing transcriptomic studies. To elucidate potential mechanisms distinguishing regenerative from non-regenerative healing, we applied spatial transcriptomics to an aging model. Consistent with other forms of repair, our digit amputation mouse model showed a significant impairment in regeneration in aged mice. Contrasting young and aged mice, spatial analysis revealed a metabolic shift in aged blastema associated with an increased bioenergetic requirement. This enhanced metabolic turnover was associated with increased hypoxia and angiogenic signaling, leading to excessive vascularization and altered regenerated bone architecture in aged mice. Administration of the metabolite oxaloacetate decreased the oxygen consumption rate of the aged blastema and increased WNT signaling, leading to enhanced in vivo bone regeneration. Thus, targeting cell metabolism may be a promising strategy to mitigate aging-induced declines in tissue regeneration.
Collapse
Affiliation(s)
- Robert J Tower
- Department of Orthopaedics, Johns Hopkins University, Baltimore, United States
| | - Emily Busse
- Department of Surgery, Tulane School of Medicine, New Orleans, United States
| | - Josue Jaramillo
- Department of Surgery, Tulane School of Medicine, New Orleans, United States
| | - Michelle Lacey
- Department of Mathematics, Tulane University, New Orleans, United States
| | - Kevin Hoffseth
- Department of Biological & Agricultural Engineering, Louisiana State University, Baton Rouge, United States
| | - Anyonya R Guntur
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, United States
| | - Jennifer Simkin
- Department of Orthopaedic Surgery, Louisiana State University Health Sciences Center, New Orleans, United States
| | - Mimi C Sammarco
- Department of Surgery, Tulane School of Medicine, New Orleans, United States
| |
Collapse
|
21
|
Lao M, Hurtado A, de Castro AC, Burgos M, Jiménez R, Barrionuevo FJ. Sox9 is required for nail bed differentiation and digit tip regeneration. J Invest Dermatol 2022; 142:2613-2622.e6. [DOI: 10.1016/j.jid.2022.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 03/15/2022] [Accepted: 03/30/2022] [Indexed: 11/28/2022]
|
22
|
Dolan CP, Imholt F, Yang TJ, Bokhari R, Gregory J, Yan M, Qureshi O, Zimmel K, Sherman KM, Falck A, Yu L, Leininger E, Brunauer R, Suva LJ, Gaddy D, Dawson LA, Muneoka K. Mouse Digit Tip Regeneration Is Mechanical Load Dependent. J Bone Miner Res 2022; 37:312-322. [PMID: 34783092 PMCID: PMC9400037 DOI: 10.1002/jbmr.4470] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 10/12/2021] [Accepted: 11/08/2021] [Indexed: 12/15/2022]
Abstract
Amputation of the mouse digit tip results in blastema-mediated regeneration. In this model, new bone regenerates de novo to lengthen the amputated stump bone, resulting in a functional replacement of the terminal phalangeal element along with associated non-skeletal tissues. Physiological examples of bone repair, such as distraction osteogenesis and fracture repair, are well known to require mechanical loading. However, the role of mechanical loading during mammalian digit tip regeneration is unknown. In this study, we demonstrate that reducing mechanical loading inhibits blastema formation by attenuating bone resorption and wound closure, resulting in the complete inhibition of digit regeneration. Mechanical unloading effects on wound healing and regeneration are completely reversible when mechanical loading is restored. Mechanical unloading after blastema formation results in a reduced rate of de novo bone formation, demonstrating mechanical load dependence of the bone regenerative response. Moreover, enhancing the wound-healing response of mechanically unloaded digits with the cyanoacrylate tissue adhesive Dermabond improves wound closure and partially rescues digit tip regeneration. Taken together, these results demonstrate that mammalian digit tip regeneration is mechanical load-dependent. Given that human fingertip regeneration shares many characteristics with the mouse digit tip, these results identify mechanical load as a previously unappreciated requirement for de novo bone regeneration in humans. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Connor P Dolan
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA.,DoD-VA Extremity Trauma and Amputation Center of Excellence, Bethesda, MD, USA.,Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Felisha Imholt
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Tae-Jung Yang
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Rihana Bokhari
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Joshua Gregory
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Mingquan Yan
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Osama Qureshi
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Katherine Zimmel
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Kirby M Sherman
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Alyssa Falck
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Ling Yu
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Eric Leininger
- Department of Cell and Molecular Biology, School of Science and Engineering, Tulane University, New Orleans, LA, USA
| | - Regina Brunauer
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Larry J Suva
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Dana Gaddy
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Lindsay A Dawson
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Ken Muneoka
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA.,Department of Cell and Molecular Biology, School of Science and Engineering, Tulane University, New Orleans, LA, USA
| |
Collapse
|
23
|
Yu L, Lin YL, Yan M, Li T, Wu EY, Zimmel K, Qureshi O, Falck A, Sherman KM, Huggins SS, Hurtado DO, Suva LJ, Gaddy D, Cai J, Brunauer R, Dawson LA, Muneoka K. Hyaline cartilage differentiation of fibroblasts in regeneration and regenerative medicine. Development 2022; 149:274141. [PMID: 35005773 PMCID: PMC8917415 DOI: 10.1242/dev.200249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/15/2021] [Indexed: 11/30/2022]
Abstract
Amputation injuries in mammals are typically non-regenerative; however, joint regeneration is stimulated by BMP9 treatment, indicating the presence of latent articular chondrocyte progenitor cells. BMP9 induces a battery of chondrogenic genes in vivo, and a similar response is observed in cultures of amputation wound cells. Extended cultures of BMP9-treated cells results in differentiation of hyaline cartilage, and single cell RNAseq analysis identified wound fibroblasts as BMP9 responsive. This culture model was used to identify a BMP9-responsive adult fibroblast cell line and a culture strategy was developed to engineer hyaline cartilage for engraftment into an acutely damaged joint. Transplanted hyaline cartilage survived engraftment and maintained a hyaline cartilage phenotype, but did not form mature articular cartilage. In addition, individual hypertrophic chondrocytes were identified in some samples, indicating that the acute joint injury site can promote osteogenic progression of engrafted hyaline cartilage. The findings identify fibroblasts as a cell source for engineering articular cartilage and establish a novel experimental strategy that bridges the gap between regeneration biology and regenerative medicine. Summary:In vivo articular cartilage regeneration serves as a model to develop novel approaches for engineering cartilage to repair damaged joints and identifies fibroblasts as a BMP9-inducible chondroprogenitor.
Collapse
Affiliation(s)
- Ling Yu
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Yu-Lieh Lin
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Mingquan Yan
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Tao Li
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, People's Republic of China
| | - Emily Y. Wu
- Dewpoint Therapeutics, 6 Tide Street, Suite 300, Boston, MA 02210, USA
| | - Katherine Zimmel
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Osama Qureshi
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Alyssa Falck
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Kirby M. Sherman
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Shannon S. Huggins
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Daniel Osorio Hurtado
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Larry J. Suva
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Dana Gaddy
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - James Cai
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Regina Brunauer
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Lindsay A. Dawson
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Ken Muneoka
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
24
|
Johnson GL, Lehoczky JA. Mammalian Digit Tip Regeneration: Moving from Phenomenon to Molecular Mechanism. Cold Spring Harb Perspect Biol 2022; 14:a040857. [PMID: 34312249 PMCID: PMC8725625 DOI: 10.1101/cshperspect.a040857] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
In this review, we present the current state of knowledge surrounding mammalian digit tip regeneration. We discuss the origin and formation of the blastema, a structure integral to digit tip regeneration, as well as recent insights driven by single-cell RNA sequencing into the molecular markers and cellular composition of the blastema. The digit tip is a composite of many different tissue types and we address what is known about the role of these separate tissues in regeneration of the whole digit tip. Specifically, we discuss the most extensively studied tissues in the digit tip: bone, nail epithelium, and peripheral nerves. We also address how known molecular pathways in limb development can inform research into digit tip regeneration. Overall, the mouse digit tip is an excellent model of complex mammalian regeneration that can provide insight into inducing regeneration in human tissues.
Collapse
Affiliation(s)
- Gemma L Johnson
- Department of Orthopedics, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Jessica A Lehoczky
- Department of Orthopedics, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| |
Collapse
|
25
|
Self-Control of Inflammation during Tail Regeneration of Lizards. J Dev Biol 2021; 9:jdb9040048. [PMID: 34842738 PMCID: PMC8629022 DOI: 10.3390/jdb9040048] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/29/2021] [Accepted: 10/31/2021] [Indexed: 12/22/2022] Open
Abstract
Lizards can spontaneously regenerate their lost tail without evoking excessive inflammation at the damaged site. In contrast, tissue/organ injury of its mammalian counterparts results in wound healing with a formation of a fibrotic scar due to uncontrolled activation of inflammatory responses. Unveiling the mechanism of self-limited inflammation occurring in the regeneration of a lizard tail will provide clues for a therapeutic alternative to tissue injury. The present review provides an overview of aspects of rapid wound healing and roles of antibacterial peptides, effects of leukocytes on the tail regeneration, self-blocking of the inflammatory activation in leukocytes, as well as inflammatory resistance of blastemal cells or immature somatic cells during lizard tail regeneration. These mechanistic insights of self-control of inflammation during lizard tail regeneration may lead in the future to the development of therapeutic strategies to fight injury-induced inflammation.
Collapse
|
26
|
Hoffseth K, Busse E, Jaramillo J, Simkin J, Lacey M, Sammarco MC. Age-Dependent Changes in Bone Architecture, Patterning, and Biomechanics During Skeletal Regeneration. Front Cell Dev Biol 2021; 9:749055. [PMID: 34722531 PMCID: PMC8548682 DOI: 10.3389/fcell.2021.749055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/22/2021] [Indexed: 12/21/2022] Open
Abstract
Mouse digit amputation provides a useful model of bone growth after injury, in that the injury promotes intramembranous bone formation in an adult animal. The digit tip is composed of skin, nerves, blood vessels, bones, and tendons, all of which regenerate after digit tip amputation, making it a powerful model for multi-tissue regeneration. Bone integrity relies upon a balanced remodeling between bone resorption and formation, which, when disrupted, results in changes to bone architecture and biomechanics, particularly during aging. In this study, we used recently developed techniques to evaluate bone patterning differences between young and aged regenerated bone. This analysis suggests that aged mice have altered trabecular spacing and patterning and increased mineral density of the regenerated bone. To further characterize the biomechanics of regenerated bone, we measured elasticity using a micro-computed tomography image-processing method combined with nanoindentation. This analysis suggests that the regenerated bone demonstrates decreased elasticity compared with the uninjured bone, but there is no significant difference in elasticity between aged and young regenerated bone. These data highlight distinct architectural and biomechanical differences in regenerated bone in both young and aged mice and provide a new analysis tool for the digit amputation model to aid in evaluating the outcomes for potential therapeutic treatments to promote regeneration.
Collapse
Affiliation(s)
- Kevin Hoffseth
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, LA, United States
| | - Emily Busse
- Department of Surgery, Tulane School of Medicine, New Orleans, LA, United States
| | - Josue Jaramillo
- Department of Surgery, Tulane School of Medicine, New Orleans, LA, United States
| | - Jennifer Simkin
- Department of Orthopaedic Surgery, Health Sciences Center, Louisiana State University, New Orleans, LA, United States
| | - Michelle Lacey
- Department of Mathematics, Tulane University, New Orleans, LA, United States
| | - Mimi C. Sammarco
- Department of Surgery, Tulane School of Medicine, New Orleans, LA, United States
| |
Collapse
|
27
|
Sader F, Roy S. Tgf-β superfamily and limb regeneration: Tgf-β to start and Bmp to end. Dev Dyn 2021; 251:973-987. [PMID: 34096672 DOI: 10.1002/dvdy.379] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/19/2022] Open
Abstract
Axolotls represent a popular model to study how nature solved the problem of regenerating lost appendages in tetrapods. Our work over many years focused on trying to understand how these animals can achieve such a feat and not end up with a scarred up stump. The Tgf-β superfamily represents an interesting family to target since they are involved in wound healing in adults and pattern formation during development. This family is large and comprises Tgf-β, Bmps, activins and GDFs. In this review, we present work from us and others on Tgf-β & Bmps and highlight interesting observations between these two sub-families. Tgf-β is important for the preparation phase of regeneration and Bmps for the redevelopment phase and they do not overlap with one another. We present novel data showing that the Tgf-β non-canonical pathway is also not active during redevelopment. Finally, we propose a molecular model to explain how Tgf-β and Bmps maintain distinct windows of expression during regeneration in axolotls.
Collapse
Affiliation(s)
- Fadi Sader
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Stéphane Roy
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada.,Department of Stomatology, Faculty of Dentistry, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
28
|
Brunauer R, Xia IG, Asrar SN, Dawson LA, Dolan CP, Muneoka K. Aging delays epimorphic regeneration in mice. J Gerontol A Biol Sci Med Sci 2021; 76:1726-1733. [PMID: 33970250 DOI: 10.1093/gerona/glab131] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Indexed: 11/14/2022] Open
Abstract
Epimorphic regeneration is a multi-tissue regeneration process where amputation does not lead to scarring, but blastema formation and patterned morphogenesis for which cell plasticity and concerted cell-cell interactions are pivotal. Tissue regeneration declines with aging, yet if and how aging impairs epimorphic regeneration is unknown. Here we show for the first time that aging derails the spatiotemporal regulation of epimorphic regeneration in mammals, first, by exacerbating tissue histolysis and delaying wound closure, and second, by impairing blastema differentiation and skeletal regrowth. Surprisingly, aging did not limit stem cell availability in the blastema, but reduced osteoblast-dependent bone formation. Our data suggest that aging delays regeneration not by stem cell exhaustion, but functional defects of differentiated cells that may be driven by an aged wound environment and alterations in the spatiotemporal regulation of regeneration events. Our findings emphasize the importance of accurate timing of signaling events for regeneration, and highlight the need for carefully timed interventions in regenerative medicine.
Collapse
Affiliation(s)
- Regina Brunauer
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Ian G Xia
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Shabistan N Asrar
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Lindsay A Dawson
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Connor P Dolan
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA.,Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, MD
| | - Ken Muneoka
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| |
Collapse
|
29
|
Guerin DJ, Kha CX, Tseng KAS. From Cell Death to Regeneration: Rebuilding After Injury. Front Cell Dev Biol 2021; 9:655048. [PMID: 33816506 PMCID: PMC8012889 DOI: 10.3389/fcell.2021.655048] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 02/22/2021] [Indexed: 12/22/2022] Open
Abstract
The ability to regrow lost or damaged tissues is widespread, but highly variable among animals. Understanding this variation remains a challenge in regeneration biology. Numerous studies from Hydra to mouse have shown that apoptosis acts as a potent and necessary mechanism in regeneration. Much is known about the involvement of apoptosis during normal development in regulating the number and type of cells in the body. In the context of regeneration, apoptosis also regulates cell number and proliferation in tissue remodeling. Apoptosis acts both early in the process to stimulate regeneration and later to regulate regenerative patterning. Multiple studies indicate that apoptosis acts as a signal to stimulate proliferation within the regenerative tissues, producing the cells needed for full regeneration. The conservation of apoptosis as a regenerative mechanism demonstrated across species highlights its importance and motivates the continued investigation of this important facet of programmed cell death. This review summarizes what is known about the roles of apoptosis during regeneration, and compares regenerative apoptosis with the mechanisms and function of apoptosis in development. Defining the complexity of regenerative apoptosis will contribute to new knowledge and perspectives for understanding mechanisms of apoptosis induction and regulation.
Collapse
Affiliation(s)
- Dylan J Guerin
- School of Life Sciences, University of Nevada, Las Vegas, Las Vegas, NV, United States
| | - Cindy X Kha
- School of Life Sciences, University of Nevada, Las Vegas, Las Vegas, NV, United States
| | - Kelly Ai-Sun Tseng
- School of Life Sciences, University of Nevada, Las Vegas, Las Vegas, NV, United States
| |
Collapse
|
30
|
Elchaninov A, Sukhikh G, Fatkhudinov T. Evolution of Regeneration in Animals: A Tangled Story. Front Ecol Evol 2021. [DOI: 10.3389/fevo.2021.621686] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The evolution of regenerative capacity in multicellular animals represents one of the most complex and intriguing problems in biology. How could such a seemingly advantageous trait as self-repair become consistently attenuated by the evolution? This review article examines the concept of the origin and nature of regeneration, its connection with the processes of embryonic development and asexual reproduction, as well as with the mechanisms of tissue homeostasis. The article presents a variety of classical and modern hypotheses explaining different trends in the evolution of regenerative capacity which is not always beneficial for the individual and notably for the species. Mechanistically, these trends are driven by the evolution of signaling pathways and progressive restriction of differentiation plasticity with concomitant advances in adaptive immunity. Examples of phylogenetically enhanced regenerative capacity are considered as well, with appropriate evolutionary reasoning for the enhancement and discussion of its molecular mechanisms.
Collapse
|
31
|
Hoffseth KF, Simkin J, Busse E, Stewart K, Watt J, Chapple A, Hargrove A, Sammarco MC. A new approach to analyzing regenerated bone quality in the mouse digit amputation model using semi-automatic processing of microCT data. Bone 2021; 144:115776. [PMID: 33276153 PMCID: PMC7906109 DOI: 10.1016/j.bone.2020.115776] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 11/04/2020] [Accepted: 11/25/2020] [Indexed: 11/25/2022]
Abstract
Bone regeneration is a critical area of research impacting treatment of diseases such as osteoporosis, age-related decline, and orthopaedic implants. A crucial question in bone regeneration is that of bone architectural quality, or how "good" is the regenerated bone tissue structurally? Current methods address typical long bone architecture, however there exists a need for improved ability to quantify structurally relevant parameters of bone in non-standard bone shapes. Here we present a new analysis approach based on open-source semi-automatic methods combining image processing, solid modeling, and numerical calculations to analyze bone tissue at a more granular level using μCT image data from a mouse digit model of bone regeneration. Examining interior architecture, growth patterning, spatial mineral content, and mineral density distribution, these methods are then applied to two types of 6-month old mouse digits - 1) those prior to amputation injury (unamputated) and 2) those 42 days after amputation when bone has regenerated. Results show regenerated digits exhibit increased inner void fraction, decreased patterning, different patterns of spatial mineral distribution, and increased mineral density values when compared to unamputated bone. Our approach demonstrates the utility of this new analysis technique in assessment of non-standard bone models, such as the regenerated bone of the digit, and aims to bring a deeper level of analysis with an open-source, integrative platform to the greater bone community.
Collapse
Affiliation(s)
- Kevin F Hoffseth
- Department of Biological & Agricultural Engineering, Louisiana State University, 149 E.B. Doran Building, Baton Rouge, LA 70803, USA.
| | - Jennifer Simkin
- Department of Orthopaedic Surgery, Louisiana State University Health Sciences Center, New Orleans, 533 Bolivar Street, New Orleans, LA 70112, USA.
| | - Emily Busse
- Department of Surgery, Tulane School of Medicine, 1400 Tulane Ave, DEPT 8622, New Orleans, LA. 70112, USA.
| | - Kennon Stewart
- Department of Surgery, Tulane School of Medicine, 1400 Tulane Ave, DEPT 8622, New Orleans, LA. 70112, USA.
| | - James Watt
- Department of Surgery, Tulane School of Medicine, 1400 Tulane Ave, DEPT 8622, New Orleans, LA. 70112, USA.
| | - Andrew Chapple
- Department of Biostatistics, Louisiana State University Health Sciences Center, New Orleans, 533 Bolivar Street, New Orleans, LA 70112, USA.
| | - Aaron Hargrove
- Department of Biological & Agricultural Engineering, Louisiana State University, 149 E.B. Doran Building, Baton Rouge, LA 70803, USA.
| | - Mimi C Sammarco
- Department of Surgery, Tulane School of Medicine, 1400 Tulane Ave, DEPT 8622, New Orleans, LA. 70112, USA.
| |
Collapse
|
32
|
Single-cell RNA-seq reveals novel mitochondria-related musculoskeletal cell populations during adult axolotl limb regeneration process. Cell Death Differ 2021; 28:1110-1125. [PMID: 33116295 PMCID: PMC7937690 DOI: 10.1038/s41418-020-00640-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 09/24/2020] [Accepted: 10/06/2020] [Indexed: 01/30/2023] Open
Abstract
While the capacity to regenerate tissues or limbs is limited in mammals, including humans, axolotls are able to regrow entire limbs and major organs after incurring a wound. The wound blastema has been extensively studied in limb regeneration. However, due to the inadequate characterization of ECM and cell subpopulations involved in the regeneration process, the discovery of the key drivers for human limb regeneration remains unknown. In this study, we applied large-scale single-cell RNA sequencing to classify cells throughout the adult axolotl limb regeneration process, uncovering a novel regeneration-specific mitochondria-related cluster supporting regeneration through energy providing and the ECM secretion (COL2+) cluster contributing to regeneration through cell-cell interactions signals. We also discovered the dedifferentiation and re-differentiation of the COL1+/COL2+ cellular subpopulation and exposed a COL2-mitochondria subcluster supporting the musculoskeletal system regeneration. On the basis of these findings, we reconstructed the dynamic single-cell transcriptome of adult axolotl limb regenerative process, and identified the novel regenerative mitochondria-related musculoskeletal populations, which yielded deeper insights into the crucial interactions between cell clusters within the regenerative microenvironment.
Collapse
|
33
|
Maturating Articular Cartilage Can Induce Ectopic Joint-Like Structures in Neonatal Mice. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2020. [DOI: 10.1007/s40883-020-00176-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Abstract
Osteoarthritis is a huge health burden to our society. Seeking for potential ways to induce regeneration of articular cartilage (AC) that is intrinsically limited, we focused on the interaction between two opposing joints. To evaluate the role of the interaction of opposing regions of AC for joint maturation, we amputated digits at the distal interphalangeal level without injuring the articular surface of the intermediate phalanx (P2) and observed that the zonal organization of AC was defective. We then removed the P2 bone without injuring the articular surface of the proximal phalanx (P1), and the remaining part of the digit was amputated near the distal interphalangeal level. The distribution pattern of type II collagen and proteoglycan 4 (PRG4) suggested that maturation of AC in P1 was delayed. These two experiments suggested that an interaction between the opposing AC in a joint is necessary for maturation of the zonal organization of AC in neonatal digits. To test if an interaction of the joints is sufficient to induce articular cartilage, a proximal fragment of P2 was resected, inverted, and put back into the original location. Newly formed cartilage was induced at the interface region between the AC of the inverted graft and the cut edge of the distal part of P2. Type II collagen and PRG4 were expressed in the ectopic cartilage in a similar manner to normal AC, indicating that neonatal AC can induce ectopic joint-like structures in mice comparable with what has been reported in newts and frogs. These results suggest that the neonatal joint could be a source of inductive signals for regeneration of AC.
Lay Summary
In this study, we experimentally show that neonatal mice appear to have the capacity to regenerate articular cartilage (AC) in digits. It is already known that mice can regenerate a digit tip after amputation, but do not regenerate in response to amputations at more proximal levels. Therefore, it has been thought that mammalian joint structures are non-regenerative. However, we found that normal digit AC can induce AC-like structures in a non-joint region when it is placed next to the cut edge of a bone, suggesting that the normal AC has regenerative capacity in certain situations in neonatal mice.
Future Works
Joint disorders are a huge health problem of our society. The results of this study suggest that neonatal AC could be a potential source of inductive signals for regeneration of AC. The discovery of these inductive signals will aid in developing regenerative therapies of a joint in human.
Collapse
|
34
|
Storer MA, Miller FD. Cellular and molecular mechanisms that regulate mammalian digit tip regeneration. Open Biol 2020; 10:200194. [PMID: 32993414 PMCID: PMC7536070 DOI: 10.1098/rsob.200194] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Digit tip regeneration is one of the few examples of true multi-tissue regeneration in an adult mammal. The key step in this process is the formation of the blastema, a transient proliferating cell mass that generates the different cell types of the digit to replicate the original structure. Failure to form the blastema results in a lack of regeneration and has been postulated to be the reason why mammalian limbs cannot regrow following amputation. Understanding how the blastema forms and functions will help us to determine what is required for mammalian regeneration to occur and will provide insights into potential therapies for mammalian tissue regeneration and repair. This review summarizes the cellular and molecular mechanisms that influence murine blastema formation and govern digit tip regeneration.
Collapse
Affiliation(s)
- Mekayla A Storer
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Canada M5G 1L7
| | - Freda D Miller
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Canada M5G 1L7.,Department of Molecular Genetics, University of Toronto, Toronto, Canada M5G 1A8.,Department of Physiology, University of Toronto, Toronto, Canada M5G 1A8.,Institute of Medical Sciences, University of Toronto, Toronto, Canada M5G 1A8
| |
Collapse
|
35
|
Dawson LA, Schanes PP, Marrero L, Jordan K, Brunauer R, Zimmel KN, Qureshi O, Imholt FM, Falck AR, Yan M, Dolan CP, Yu L, Muneoka K. Proximal digit tip amputation initiates simultaneous blastema and transient fibrosis formation and results in partial regeneration. Wound Repair Regen 2020; 29:196-205. [PMID: 32815252 DOI: 10.1111/wrr.12856] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/09/2020] [Accepted: 07/23/2020] [Indexed: 12/14/2022]
Abstract
Complete extremity regeneration in mammals is restricted to distal amputations of the digit tip, the terminal phalanx (P3). In mice, P3 regeneration is mediated via the formation of a blastema, a transient population of progenitor cells that form from the blending of periosteal and endosteal/marrow compartmentalized cells that undergo differentiation to restore the amputated structures. Compartmentalized blastema cells are formed independently, and periosteal compartment-derived cells are required for restoration of amputated skeletal length. P3 regenerative capacity is progressively attenuated at increasingly more proximal amputation levels, eventually resulting in regenerative failure. The continuum of regenerative capacity within the P3 wound milieu is a unique model to investigate mammalian blastema formation in response to distal amputation, as well as the healing response associated with regenerative failure at proximal amputation levels. We report that P3 proximal amputation healing, previously reported to result in regenerative failure, is not an example of complete regenerative failure, but instead is characterized by a limited bone regeneration response restricted to the endosteal/marrow compartment. The regeneration response is mediated by blastema formation within the endosteal/marrow compartment, and blastemal osteogenesis progresses through intramembranous ossification in a polarized proximal to distal sequence. Unlike bone regeneration following distal P3 amputation, osteogenesis within the periosteal compartment is not observed in response to proximal P3 amputation. We provide evidence that proximal P3 amputation initiates the formation of fibrotic tissue that isolates the endosteal/marrow compartment from the periosteal compartment and wound epidermis. While the fibrotic response is transient and later resolved, these studies demonstrate that blastema formation and fibrosis can occur in close proximity, with the regenerative response dominating the final outcome. Moreover, the results suggest that the attenuated proximal P3 regeneration response is associated with the absence of periosteal-compartment participation in blastema formation and bone regeneration.
Collapse
Affiliation(s)
- Lindsay A Dawson
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Paula P Schanes
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana, USA
| | - Luis Marrero
- Department of Orthopedic Surgery, Louisiana State University School of Medicine, New Orleans, Louisiana, USA.,Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Kathryn Jordan
- Department of Orthopedic Surgery, Louisiana State University School of Medicine, New Orleans, Louisiana, USA.,Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.,College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Regina Brunauer
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Katherine N Zimmel
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Osama Qureshi
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Felisha M Imholt
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Alyssa R Falck
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Mingquan Yan
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Connor P Dolan
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Ling Yu
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Ken Muneoka
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
36
|
Qu F, Palte IC, Gontarz PM, Zhang B, Guilak F. Transcriptomic analysis of bone and fibrous tissue morphogenesis during digit tip regeneration in the adult mouse. FASEB J 2020; 34:9740-9754. [PMID: 32506623 DOI: 10.1096/fj.202000330r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/29/2020] [Accepted: 05/15/2020] [Indexed: 12/31/2022]
Abstract
Humans have limited regenerative potential of musculoskeletal tissues following limb or digit loss. The murine digit has been used to study mammalian regeneration, where stem/progenitor cells (the "blastema") completely regenerate the digit tip after distal, but not proximal, amputation. However, the molecular mechanisms responsible for this response remain to be determined. Here, we evaluated the spatiotemporal formation of bone and fibrous tissues after level-dependent amputation of the murine terminal phalanx and quantified the transcriptome of the repair tissue. Distal (regenerative) and proximal (non-regenerative) amputations showed significant differences in temporal gene expression and tissue regrowth over time. Genes that direct skeletal system development and limb morphogenesis are transiently upregulated during blastema formation and differentiation, including distal Hox genes. Overall, our results suggest that digit tip regeneration is controlled by a gene regulatory network that recapitulates aspects of limb development, and that failure to activate this developmental program results in fibrotic wound healing.
Collapse
Affiliation(s)
- Feini Qu
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA.,Center of Regenerative Medicine, Washington University, St. Louis, MO, USA.,Shriners Hospitals for Children-St. Louis, St. Louis, MO, USA
| | - Ilan C Palte
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA.,Center of Regenerative Medicine, Washington University, St. Louis, MO, USA.,Shriners Hospitals for Children-St. Louis, St. Louis, MO, USA
| | - Paul M Gontarz
- Center of Regenerative Medicine, Washington University, St. Louis, MO, USA
| | - Bo Zhang
- Center of Regenerative Medicine, Washington University, St. Louis, MO, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO, USA.,Center of Regenerative Medicine, Washington University, St. Louis, MO, USA.,Shriners Hospitals for Children-St. Louis, St. Louis, MO, USA
| |
Collapse
|
37
|
Papalois ZA, Papalois KB. Bioethics and Environmental Ethics: The Story of the Human Body as a Natural Ecosystem. New Bioeth 2020; 26:91-97. [PMID: 32597380 DOI: 10.1080/20502877.2020.1767919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Is there a parallel between climate change and our body's temperature or non-compliance and failure to act on global warming? This paper proposes a model which describes the human body as part of Nature's ecosystem. By utilising the power of observation to identify a problem, environmental and applied ethics can guide action and instigate change, not only to change the predicted plot of climate change, but also the wellbeing of humans in life's story. Through a discussion on human autonomy and lessons learned from the past, earth's inhabitants can identify a balance between beneficence and non-maleficence for themselves and our planet.
Collapse
|
38
|
Yu Y, Cui H, Zhang C, Zhang D, Yin J, Wen G, Chai Y. Human nail bed extracellular matrix facilitates bone regeneration via macrophage polarization mediated by the JAK2/STAT3 pathway. J Mater Chem B 2020; 8:4067-4079. [PMID: 32242565 DOI: 10.1039/c9tb02910a] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Critical-sized bone defects caused by trauma, tumor resection or serious infection represent one of the most challenging problems faced by orthopedic surgeons. However, the construction of bone grafts with good osteointegration and osteoinductivity is a clinical challenge. It has been elaborated that the nail bed tissue is an essential element for digit tip regeneration, suggesting that the nail bed may serve as a new material to manipulate bone regeneration. Herein, it was found that human nail bed extracellular matrix derived from amputated patients stimulates macrophage polarization toward a pro-healing phenotype and the expression of BMP2, to facilitate the osteogenic differentiation of bone marrow stromal cells (BMSCs) in vitro. The in vivo osteogenic capacity of decellularized nail bed scaffolds was then confirmed using a rat model of critical-sized calvarial defects. The in-depth analysis of immune responses to implanted scaffolds revealed that macrophage polarization toward the pro-regenerative M2 phenotype directs osteogenesis, as confirmed by macrophage depletion. A combination of proteomics analysis and RNA interference verified that the JAK2/STAT3 pathway is the positive regulator of macrophage polarization initiated by the decellularized nail bed during the promoted osteogenesis process. Thus, the decellularized human nail bed scaffold developed in this work is a promising biomaterial for bone regeneration.
Collapse
Affiliation(s)
- Yaling Yu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.
| | | | | | | | | | | | | |
Collapse
|
39
|
Kaji DA, Tan Z, Johnson GL, Huang W, Vasquez K, Lehoczky JA, Levi B, Cheah KS, Huang AH. Cellular Plasticity in Musculoskeletal Development, Regeneration, and Disease. J Orthop Res 2020; 38:708-718. [PMID: 31721278 PMCID: PMC7213644 DOI: 10.1002/jor.24523] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 11/04/2019] [Indexed: 02/04/2023]
Abstract
In this review, we highlight themes from a recent workshop focused on "Plasticity of Cell Fate in Musculoskeletal Tissues" held at the Orthopaedic Research Society's 2019 annual meeting. Experts in the field provided examples of mesenchymal cell plasticity during normal musculoskeletal development, regeneration, and disease. A thorough understanding of the biology underpinning mesenchymal cell plasticity may offer a roadmap for promoting regeneration while attenuating pathologic differentiation. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:708-718, 2020.
Collapse
Affiliation(s)
- Deepak A. Kaji
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, NYC, NY, USA
| | - Zhijia Tan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong
| | - Gemma L. Johnson
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Boston, MA, USA,Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Wesley Huang
- Department of Plastic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Kaetlin Vasquez
- Department of Plastic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Jessica A. Lehoczky
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Boston, MA, USA
| | - Benjamin Levi
- Department of Plastic Surgery, University of Michigan, Ann Arbor, MI, USA
| | | | - Alice H. Huang
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, NYC, NY, USA
| |
Collapse
|
40
|
Ritschka B, Knauer-Meyer T, Gonçalves DS, Mas A, Plassat JL, Durik M, Jacobs H, Pedone E, Di Vicino U, Cosma MP, Keyes WM. The senotherapeutic drug ABT-737 disrupts aberrant p21 expression to restore liver regeneration in adult mice. Genes Dev 2020; 34:489-494. [PMID: 32139422 PMCID: PMC7111259 DOI: 10.1101/gad.332643.119] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 02/04/2020] [Indexed: 12/15/2022]
Abstract
In this study, Ritschka et al. investigated whether cellular senescence might play a role in loss of regenerative capacity during liver regeneration. They show that following partial hepatectomy, the senescence-associated genes p21, p16Ink4a, and p19Arf become dynamically expressed in different cell types when regenerative capacity decreases, but without a full senescent response, and that treatment with a senescence-inhibiting drug improves regeneration through targeting aberrantly prolonged p21 expression. Young mammals possess a limited regenerative capacity in some tissues, which is lost upon maturation. We investigated whether cellular senescence might play a role in such loss during liver regeneration. We found that following partial hepatectomy, the senescence-associated genes p21, p16Ink4a, and p19Arf become dynamically expressed in different cell types when regenerative capacity decreases, but without a full senescent response. However, we show that treatment with a senescence-inhibiting drug improves regeneration, by disrupting aberrantly prolonged p21 expression. This work suggests that senescence may initially develop from heterogeneous cellular responses, and that senotherapeutic drugs might be useful in promoting organ regeneration.
Collapse
Affiliation(s)
- Birgit Ritschka
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch 67404, France.,UMR7104, Centre National de la Recherche Scientifique (CNRS), Illkirch 67404, France.,U1258, Institut National de la Santé et de la Recherche Médicale (INSERM), Illkirch 67404, France.,Université de Strasbourg, Illkirch 67404, France.,Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona 08003, Spain
| | - Tania Knauer-Meyer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch 67404, France.,UMR7104, Centre National de la Recherche Scientifique (CNRS), Illkirch 67404, France.,U1258, Institut National de la Santé et de la Recherche Médicale (INSERM), Illkirch 67404, France.,Université de Strasbourg, Illkirch 67404, France
| | - Daniel Sampaio Gonçalves
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch 67404, France.,UMR7104, Centre National de la Recherche Scientifique (CNRS), Illkirch 67404, France.,U1258, Institut National de la Santé et de la Recherche Médicale (INSERM), Illkirch 67404, France.,Université de Strasbourg, Illkirch 67404, France
| | - Alba Mas
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch 67404, France.,UMR7104, Centre National de la Recherche Scientifique (CNRS), Illkirch 67404, France.,U1258, Institut National de la Santé et de la Recherche Médicale (INSERM), Illkirch 67404, France.,Université de Strasbourg, Illkirch 67404, France.,Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona 08003, Spain
| | - Jean-Luc Plassat
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch 67404, France.,UMR7104, Centre National de la Recherche Scientifique (CNRS), Illkirch 67404, France.,U1258, Institut National de la Santé et de la Recherche Médicale (INSERM), Illkirch 67404, France.,Université de Strasbourg, Illkirch 67404, France
| | - Matej Durik
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch 67404, France.,UMR7104, Centre National de la Recherche Scientifique (CNRS), Illkirch 67404, France.,U1258, Institut National de la Santé et de la Recherche Médicale (INSERM), Illkirch 67404, France.,Université de Strasbourg, Illkirch 67404, France
| | - Hugues Jacobs
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch 67404, France.,UMR7104, Centre National de la Recherche Scientifique (CNRS), Illkirch 67404, France.,U1258, Institut National de la Santé et de la Recherche Médicale (INSERM), Illkirch 67404, France.,Université de Strasbourg, Illkirch 67404, France
| | - Elisa Pedone
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona 08003, Spain
| | - Umberto Di Vicino
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona 08003, Spain
| | - Maria Pia Cosma
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona 08003, Spain.,Universitat Pompeu Fabra (UPF), Barcelona 08003, Spain.,Institución Catalana de Investigación y Estudios Avanzados (ICREA), Barcelona 08010, Spain
| | - William M Keyes
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch 67404, France.,UMR7104, Centre National de la Recherche Scientifique (CNRS), Illkirch 67404, France.,U1258, Institut National de la Santé et de la Recherche Médicale (INSERM), Illkirch 67404, France.,Université de Strasbourg, Illkirch 67404, France.,Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona 08003, Spain.,Universitat Pompeu Fabra (UPF), Barcelona 08003, Spain
| |
Collapse
|
41
|
Transcriptional analysis of scar-free wound healing during early stages of tail regeneration in the green anole lizard, Anolis carolinensis. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/j.regen.2019.100025] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
42
|
Johnson GL, Masias EJ, Lehoczky JA. Cellular Heterogeneity and Lineage Restriction during Mouse Digit Tip Regeneration at Single-Cell Resolution. Dev Cell 2020; 52:525-540.e5. [PMID: 32097654 PMCID: PMC7186907 DOI: 10.1016/j.devcel.2020.01.026] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 12/27/2022]
Abstract
Innate regeneration following digit tip amputation is one of the few examples of epimorphic regeneration in mammals. Digit tip regeneration is mediated by the blastema, the same structure invoked during limb regeneration in some lower vertebrates. By genetic lineage analyses, the digit tip blastema has been defined as a population of heterogeneous, lineage-restricted progenitor cells. These previous studies, however, do not comprehensively evaluate blastema heterogeneity or address lineage restriction of closely related cell types. In this report, we present single-cell RNA sequencing of over 38,000 cells from mouse digit tip blastemas and unamputated control digit tips and generate an atlas of the cell types participating in digit tip regeneration. We computationally define differentiation trajectories of vascular, monocytic, and fibroblastic lineages over regeneration, and while our data confirm broad lineage restriction of progenitors, our analysis reveals 67 genes enriched in blastema fibroblasts including a novel regeneration-specific gene, Mest.
Collapse
Affiliation(s)
- Gemma L Johnson
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Erick J Masias
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Jessica A Lehoczky
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
43
|
Muneoka K, Dawson LA. Evolution of epimorphosis in mammals. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2020; 336:165-179. [PMID: 31951104 DOI: 10.1002/jez.b.22925] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 10/29/2019] [Accepted: 12/23/2019] [Indexed: 12/30/2022]
Abstract
Mammalian epimorphic regeneration is rare and digit tip regeneration in mice is the best-studied model for a multi-tissue regenerative event that involves blastema formation. Digit tip regeneration parallels human fingertip regeneration, thus understanding the details of this response can provide insight into developing strategies to expand the potential of human regeneration. Following amputation, the digit stump undergoes a strong histolytic response involving osteoclast-mediated bone degradation that is spatially and temporally linked to the expansion of blastema osteoprogenitor cells. Blastemal differentiation occurs via direct intramembranous ossification. Although robust, digit regeneration is imperfect: The amputated cortical bone is replaced with woven bone and there is excessive bone regeneration restricted to the dorsal-ventral axis. Ontogenetic and phylogenetic analysis of digit regeneration in amphibians and mammals raise the possibility that mammalian blastema is a product of convergent evolution and we hypothesize that digit tip regeneration evolved from a nonregenerative precondition. A model is proposed in which the mammalian blastema evolved in part from an adaptation of two bone repair strategies (the bone remodeling cycle and fracture healing) both of which are conserved across tetrapod vertebrates. The view that epimorphic regeneration evolved in mammals from a nonregenerative precondition is supported by recent studies demonstrating that complex regenerative responses can be induced from a number of different nonregenerative amputation wounds by specific modification of the healing response.
Collapse
Affiliation(s)
- Ken Muneoka
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Lindsay A Dawson
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| |
Collapse
|
44
|
Storer MA, Mahmud N, Karamboulas K, Borrett MJ, Yuzwa SA, Gont A, Androschuk A, Sefton MV, Kaplan DR, Miller FD. Acquisition of a Unique Mesenchymal Precursor-like Blastema State Underlies Successful Adult Mammalian Digit Tip Regeneration. Dev Cell 2020; 52:509-524.e9. [PMID: 31902657 DOI: 10.1016/j.devcel.2019.12.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 11/11/2019] [Accepted: 12/09/2019] [Indexed: 12/11/2022]
Abstract
Here, we investigate the origin and nature of blastema cells that regenerate the adult murine digit tip. We show that Pdgfra-expressing mesenchymal cells in uninjured digits establish the regenerative blastema and are essential for regeneration. Single-cell profiling shows that the mesenchymal blastema cells are distinct from both uninjured digit and embryonic limb or digit Pdgfra-positive cells. This unique blastema state is environmentally determined; dermal fibroblasts transplanted into the regenerative, but not non-regenerative, digit express blastema-state genes and contribute to bone regeneration. Moreover, lineage tracing with single-cell profiling indicates that endogenous osteoblasts or osteocytes acquire a blastema mesenchymal transcriptional state and contribute to both dermis and bone regeneration. Thus, mammalian digit tip regeneration occurs via a distinct adult mechanism where the regenerative environment promotes acquisition of a blastema state that enables cells from tissues such as bone to contribute to the regeneration of other mesenchymal tissues such as the dermis.
Collapse
Affiliation(s)
- Mekayla A Storer
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto M5G 1L7, Canada
| | - Neemat Mahmud
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto M5G 1A8, Canada
| | - Konstantina Karamboulas
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto M5G 1L7, Canada
| | - Michael J Borrett
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto M5G 1L7, Canada; Institute of Medical Sciences, University of Toronto, Toronto M5G 1A8, Canada
| | - Scott A Yuzwa
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto M5G 1L7, Canada
| | - Alexander Gont
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto M5G 1L7, Canada
| | - Alaura Androschuk
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto M5G 1A8, Canada
| | - Michael V Sefton
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto M5G 1A8, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto M5G 1A8, Canada
| | - David R Kaplan
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto M5G 1A8, Canada; Institute of Medical Sciences, University of Toronto, Toronto M5G 1A8, Canada
| | - Freda D Miller
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto M5G 1A8, Canada; Department of Physiology, University of Toronto, Toronto M5G 1A8, Canada; Institute of Medical Sciences, University of Toronto, Toronto M5G 1A8, Canada.
| |
Collapse
|
45
|
Lu S, Xiong Q, Du K, Gan X, Wang X, Yang L, Wang Y, Ge F, He S. Comparative iTRAQ proteomics revealed proteins associated with lobed fin regeneration in Bichirs. Proteome Sci 2019; 17:6. [PMID: 31832023 PMCID: PMC6869209 DOI: 10.1186/s12953-019-0153-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 10/09/2019] [Indexed: 11/23/2022] Open
Abstract
Background Polypterus senegalus can fully regenerate its pectoral lobed fins, including a complex endoskeleton, with remarkable precision. However, despite the enormous potential of this species for use in medical research, its regeneration mechanisms remain largely unknown. Methods To identify the differentially expressed proteins (DEPs) during the early stages of lobed fin regeneration in P. senegalus, we performed a differential proteomic analysis using isobaric tag for relative and absolute quantitation (iTRAQ) approach based quantitative proteome from the pectoral lobed fins at 3 time points. Furthermore, we validated the changes in protein expression with multiple-reaction monitoring (MRM) analysis. Results The experiment yielded a total of 3177 proteins and 15,091 unique peptides including 1006 non-redundant (nr) DEPs. Of these, 592 were upregulated while 349 were downregulated after lobed fin amputation when compared to the original tissue. Bioinformatics analyses showed that the DEPs were mainly associated with Ribosome and RNA transport, metabolic, ECM-receptor interaction, Golgi and endoplasmic reticulum, DNA replication, and Regulation of actin cytoskeleton. Conclusions To our knowledge, this is the first proteomic research to investigate alterations in protein levels and affected pathways in bichirs’ lobe-fin/limb regeneration. In addition, our study demonstrated a highly dynamic regulation during lobed fin regeneration in P. senegalus. These results not only provide a comprehensive dataset on differentially expressed proteins during the early stages of lobe-fin/limb regeneration but also advance our understanding of the molecular mechanisms underlying lobe-fin/limb regeneration.
Collapse
Affiliation(s)
- Suxiang Lu
- 1Key Laboratory of Aquatic Biodiversity and Conservation of the Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072 Hubei China.,2Present address: Medical College of Pingdingshan University, Pingdingshan, 467000 Henan Province China
| | - Qian Xiong
- 3Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072 Hubei China
| | - Kang Du
- 1Key Laboratory of Aquatic Biodiversity and Conservation of the Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072 Hubei China
| | - Xiaoni Gan
- 1Key Laboratory of Aquatic Biodiversity and Conservation of the Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072 Hubei China
| | - Xuzhen Wang
- 1Key Laboratory of Aquatic Biodiversity and Conservation of the Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072 Hubei China
| | - Liandong Yang
- 1Key Laboratory of Aquatic Biodiversity and Conservation of the Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072 Hubei China
| | - Ying Wang
- 1Key Laboratory of Aquatic Biodiversity and Conservation of the Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072 Hubei China
| | - Feng Ge
- 3Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072 Hubei China
| | - Shunping He
- 1Key Laboratory of Aquatic Biodiversity and Conservation of the Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072 Hubei China
| |
Collapse
|
46
|
Easterling MR, Engbrecht KM, Crespi EJ. Endocrine Regulation of Epimorphic Regeneration. Endocrinology 2019; 160:2969-2980. [PMID: 31593236 DOI: 10.1210/en.2019-00321] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 10/01/2019] [Indexed: 12/16/2022]
Abstract
Studies aiming to uncover primary mechanisms of regeneration have predominantly focused on genetic pathways regulating specific stages in the regeneration process: wound healing, blastema formation, and pattern formation. However, studies across organisms show that environmental conditions and the physiological state of the animal can affect the rate or quality of regeneration, and endocrine signals are likely the mediators of these effects. Endocrine signals acting directly on receptors expressed in the tissue or via neuroendocrine pathways can affect regeneration by regulating the immune response to injury, allocation of energetic resources, or by enhancing or inhibiting proliferation and differentiation pathways involved in regeneration. This review discusses the cumulative knowledge in the literature about endocrine regulation of regeneration and its importance in future research to advance biomedical research.
Collapse
Affiliation(s)
- Marietta R Easterling
- School of Biological Sciences, Center for Reproductive Biology, Washington State University, Pullman, Washington
| | - Kristin M Engbrecht
- School of Biological Sciences, Center for Reproductive Biology, Washington State University, Pullman, Washington
- Pacific Northwest National Laboratory, Richland, Washington
| | - Erica J Crespi
- School of Biological Sciences, Center for Reproductive Biology, Washington State University, Pullman, Washington
| |
Collapse
|
47
|
Sun X, Gao X, Deng Z, Zhang L, McGilvray K, Gadomski BC, Amra S, Bao G, Huard J. High bone microarchitecture, strength, and resistance to bone loss in MRL/MpJ mice correlates with activation of different signaling pathways and systemic factors. FASEB J 2019; 34:789-806. [PMID: 31914651 DOI: 10.1096/fj.201901229rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 10/30/2019] [Accepted: 11/02/2019] [Indexed: 12/27/2022]
Abstract
The MRL/MpJ mice have demonstrated an enhanced tissue regeneration capacity for various tissues. In the present study, we systematically characterized bone microarchitecture and found that MRL/MpJ mice exhibit higher bone microarchitecture and strength compared to both C57BL/10J and C57BL/6J WT mice at 2, 4, and 10 months of age. The higher bone mass in MRL/MpJ mice was correlated to increased osteoblasts, decreased osteoclasts, higher cell proliferation, and bone formation, and enhanced pSMAD5 signaling earlier during postnatal development (2-month old) in the spine trabecular bone, and lower bone resorption rate at later age. Furthermore, these mice exhibit accelerated fracture healing via enhanced pSMAD5, pAKT and p-P38MAPK pathways compared to control groups. Moreover, MRL/MpJ mice demonstrated resistance to ovariectomy-induced bone loss as evidenced by maintaining higher bone volume/tissue volume (BV/TV) and lower percentage of bone loss later after ovariectomy. The consistently higher serum IGF1 level and lower RANKL level in MRL/MpJ mice may contribute to the maintenance of high bone mass in uninjured and injured bone. In conclusion, our results indicate that enhanced pSMAD5, pAKT, and p-P38MAPK signaling, higher serum IGF-1, and lower RANKL level contribute to the higher bone microarchitecture and strength, accelerated healing, and resistance to osteoporosis in MRL/MpJ mice.
Collapse
Affiliation(s)
- Xuying Sun
- Department of Orthopaedic Surgery, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Xueqin Gao
- Department of Orthopaedic Surgery, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas.,Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, Colorado
| | - Zhenhan Deng
- Department of Orthopaedic Surgery, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Linlin Zhang
- Department of Biomedical Engineering, Rice University, Houston, Texas
| | - Kirk McGilvray
- Orthopaedic Bioengineering Research Laboratory, Department of Mechanical Engineering and School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado
| | - Benjamin C Gadomski
- Orthopaedic Bioengineering Research Laboratory, Department of Mechanical Engineering and School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado
| | - Sarah Amra
- Department of Orthopaedic Surgery, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Gang Bao
- Department of Biomedical Engineering, Rice University, Houston, Texas
| | - Johnny Huard
- Department of Orthopaedic Surgery, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas.,Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, Colorado
| |
Collapse
|
48
|
Sirtuin 3 deficiency does not impede digit regeneration in mice. Sci Rep 2019; 9:16491. [PMID: 31712596 PMCID: PMC6848098 DOI: 10.1038/s41598-019-52921-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 10/25/2019] [Indexed: 12/11/2022] Open
Abstract
The mitochondrial deacetylase sirtuin 3 (SIRT3) is thought to be one of the main contributors to metabolic flexibility-promoting mitochondrial energy production and maintaining homeostasis. In bone, metabolic profiles are tightly regulated and the loss of SIRT3 has deleterious effects on bone volume in vivo and on osteoblast differentiation in vitro. Despite the prominent role of this protein in bone stem cell proliferation, metabolic activity, and differentiation, the importance of SIRT3 for regeneration after bone injury has never been reported. We show here, using the mouse digit amputation model, that SIRT3 deficiency has no impact on the regenerative capacity and architecture of bone and soft tissue. Regeneration occurs in SIRT3 deficient mice in spite of the reduced oxidative metabolic profile of the periosteal cells. These data suggest that bone regeneration, in contrast to homeostatic bone turnover, is not reliant upon active SIRT3, and our results highlight the need to examine known roles of SIRT3 in the context of injury.
Collapse
|
49
|
Stierli S, Imperatore V, Lloyd AC. Schwann cell plasticity-roles in tissue homeostasis, regeneration, and disease. Glia 2019; 67:2203-2215. [PMID: 31215712 DOI: 10.1002/glia.23643] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/14/2019] [Accepted: 05/14/2019] [Indexed: 12/12/2022]
Abstract
How tissues are maintained over a lifetime and repaired following injury are fundamental questions in biology with a disruption to these processes underlying pathologies such as cancer and degenerative disorders. It is becoming increasingly clear that each tissue has a distinct mechanism to maintain homeostasis and respond to injury utilizing different types of stem/progenitor cell populations depending on the insult and/or with a contribution from more differentiated cells that are able to dedifferentiate to aid tissue regeneration. Peripheral nerves are highly quiescent yet show remarkable regenerative capabilities. Remarkably, there is no evidence for a classical stem cell population, rather all cell-types within the nerve are able to proliferate to produce new nerve tissue. Co-ordinating the regeneration of this tissue are Schwann cells (SCs), the main glial cells of the peripheral nervous system. SCs exist in architecturally stable structures that can persist for the lifetime of an animal, however, they are not postmitotic, in that following injury they are reprogrammed at high efficiency to a progenitor-like state, with these cells acting to orchestrate the nerve regeneration process. During nerve regeneration, SCs show little plasticity, maintaining their identity in the repaired tissue. However, once free of the nerve environment they appear to exhibit increased plasticity with reported roles in the repair of other tissues. In this review, we will discuss the mechanisms underlying the homeostasis and regeneration of peripheral nerves and how reprogrammed progenitor-like SCs have broader roles in the repair of other tissues with implications for pathologies such as cancer.
Collapse
Affiliation(s)
- Salome Stierli
- MRC LMCB, University College London, Gower Street, London, WC1E 6BT, UK
| | | | - Alison C Lloyd
- MRC LMCB, University College London, Gower Street, London, WC1E 6BT, UK
| |
Collapse
|
50
|
Easterling MR, Engbrecht KM, Crespi EJ. Endocrine regulation of regeneration: Linking global signals to local processes. Gen Comp Endocrinol 2019; 283:113220. [PMID: 31310748 DOI: 10.1016/j.ygcen.2019.113220] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/08/2019] [Accepted: 07/08/2019] [Indexed: 01/10/2023]
Abstract
Regeneration in amphibians and reptiles has been explored since the early 18th century, giving us a working in vivo model to study epimorphic regeneration in vertebrates. Studies aiming to uncover primary mechanisms of regeneration have predominantly focused on genetic pathways regulating specific stages of the regeneration process: wound healing, blastema formation and growth, and pattern formation. However, studies across organisms show that environmental conditions and physiological state of the animal can affect the rate or quality of regeneration, and endocrine signals are likely the mediators of these effects. Endocrine signals working/acting directly on receptors expressed in the structure or via neuroendocrine pathways can affect regeneration by modulating immune response to injury, allocation of energetic resources, or by enhancing or inhibiting proliferation and differentiation pathways in regenerating tissue. This review discusses the cumulative knowledge known about endocrine regulation of regeneration and important future research directions of interest to both ecological and biomedical research.
Collapse
Affiliation(s)
- Marietta R Easterling
- Washington State University, School of Biological Sciences, Center for Reproductive Biology, Pullman, WA 99164, United States.
| | - Kristin M Engbrecht
- Washington State University, School of Biological Sciences, Center for Reproductive Biology, Pullman, WA 99164, United States; Pacific Northwest National Laboratory, Richland, WA 99352, United States
| | - Erica J Crespi
- Washington State University, School of Biological Sciences, Center for Reproductive Biology, Pullman, WA 99164, United States
| |
Collapse
|