1
|
Imere A, Foster NC, Hajiali H, Okur KE, Wright AL, Barroso IA, Haj AJE. Enhanced chondrogenic potential in GelMA-based 3D cartilage model via Wnt3a surface immobilization. Sci Rep 2024; 14:15022. [PMID: 38951570 PMCID: PMC11217376 DOI: 10.1038/s41598-024-65970-w] [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: 03/12/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024] Open
Abstract
Cartilage tissue engineering aims to develop functional substitutes for treating cartilage defects and osteoarthritis. Traditional two-dimensional (2D) cell culture systems lack the complexity of native cartilage, leading to the development of 3D regenerative cartilage models. In this study, we developed a 3D model using Gelatin Methacryloyl (GelMA)-based hydrogels seeded with Y201 cells, a bone marrow mesenchymal stem cell line. The model investigated chondrogenic differentiation potential in response to Wnt3a stimulation within the GelMA scaffold and validated using known chondrogenic agonists. Y201 cells demonstrated suitability for the model, with increased proteoglycan content and upregulated chondrogenic marker expression under chondrogenic conditions. Wnt3a enhanced cell proliferation, indicating activation of the Wnt/β-catenin pathway, which plays a role in cartilage development. GelMA hydrogels provided an optimal scaffold, supporting cell viability and proliferation. The 3D model exhibited consistent responses to chondrogenic agonists, with TGF-β3 enhancing cartilage-specific extracellular matrix (ECM) production and chondrogenic differentiation. The combination of Wnt3a and TGF-β3 showed synergistic effects, promoting chondrogenic differentiation and ECM production. This study presents a 3D regenerative cartilage model with potential for investigating cartilage biology, disease mechanisms, and drug screening. The model provides insights into complex cartilage regeneration mechanisms and offers a platform for developing therapeutic approaches for cartilage repair and osteoarthritis treatment.
Collapse
Affiliation(s)
- Angela Imere
- Healthcare Technologies Institute, Institute of Translational Medicine, National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK
| | - Nicola C Foster
- Healthcare Technologies Institute, Institute of Translational Medicine, National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK
| | - Hadi Hajiali
- Healthcare Technologies Institute, Institute of Translational Medicine, National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK.
| | - Kerime Ebrar Okur
- Healthcare Technologies Institute, Institute of Translational Medicine, National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK
| | - Abigail L Wright
- Healthcare Technologies Institute, Institute of Translational Medicine, National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK
| | - Ines A Barroso
- Healthcare Technologies Institute, Institute of Translational Medicine, National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK
| | - Alicia J El Haj
- Healthcare Technologies Institute, Institute of Translational Medicine, National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
2
|
Bedelbaeva K, Cameron B, Latella J, Aslanukov A, Gourevitch D, Davuluri R, Heber-Katz E. Epithelial-mesenchymal transition: an organizing principle of mammalian regeneration. Front Cell Dev Biol 2023; 11:1101480. [PMID: 37965571 PMCID: PMC10641390 DOI: 10.3389/fcell.2023.1101480] [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: 11/17/2022] [Accepted: 09/27/2023] [Indexed: 11/16/2023] Open
Abstract
Introduction: The MRL mouse strain is one of the few examples of a mammal capable of healing appendage wounds by regeneration, a process that begins with the formation of a blastema, a structure containing de-differentiating mesenchymal cells. HIF-1α expression in the nascent MRL wound site blastema is one of the earliest identified events and is sufficient to initiate the complete regenerative program. However, HIF-1α regulates many cellular processes modulating the expression of hundreds of genes. A later signal event is the absence of a functional G1 checkpoint, leading to G2 cell cycle arrest with increased cellular DNA but little cell division observed in the blastema. This lack of mitosis in MRL blastema cells is also a hallmark of regeneration in classical invertebrate and vertebrate regenerators such as planaria, hydra, and newt. Results and discussion: Here, we explore the cellular events occurring between HIF-1α upregulation and its regulation of the genes involved in G2 arrest (EVI-5, γH3, Wnt5a, and ROR2), and identify epithelial-mesenchymal transition (EMT) (Twist and Slug) and chromatin remodeling (EZH-2 and H3K27me3) as key intermediary processes. The locus of these cellular events is highly regionalized within the blastema, occurring in the same cells as determined by double staining by immunohistochemistry and FACS analysis, and appears as EMT and chromatin remodeling, followed by G2 arrest determined by kinetic expression studies.
Collapse
Affiliation(s)
- Kamila Bedelbaeva
- Lankenau Institute for Medical Research (LIMR), Wynnewood, PA, United States
| | - Benjamin Cameron
- Lankenau Institute for Medical Research (LIMR), Wynnewood, PA, United States
| | - John Latella
- Lankenau Institute for Medical Research (LIMR), Wynnewood, PA, United States
| | - Azamat Aslanukov
- Lankenau Institute for Medical Research (LIMR), Wynnewood, PA, United States
| | | | | | - Ellen Heber-Katz
- Lankenau Institute for Medical Research (LIMR), Wynnewood, PA, United States
- The Wistar Institute, Philadelphia, PA, United States
| |
Collapse
|
3
|
Mack KL, Talbott HE, Griffin MF, Parker JBL, Guardino NJ, Spielman AF, Davitt MF, Mascharak S, Downer M, Morgan A, Valencia C, Akras D, Berger MJ, Wan DC, Fraser HB, Longaker MT. Allele-specific expression reveals genetic drivers of tissue regeneration in mice. Cell Stem Cell 2023; 30:1368-1381.e6. [PMID: 37714154 PMCID: PMC10592051 DOI: 10.1016/j.stem.2023.08.010] [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: 08/05/2022] [Revised: 06/16/2023] [Accepted: 08/22/2023] [Indexed: 09/17/2023]
Abstract
In adult mammals, skin wounds typically heal by scarring rather than through regeneration. In contrast, "super-healer" Murphy Roths Large (MRL) mice have the unusual ability to regenerate ear punch wounds; however, the molecular basis for this regeneration remains elusive. Here, in hybrid crosses between MRL and non-regenerating mice, we used allele-specific gene expression to identify cis-regulatory variation associated with ear regeneration. Analyzing three major cell populations (immune, fibroblast, and endothelial), we found that genes with cis-regulatory differences specifically in fibroblasts were associated with wound-healing pathways and also co-localized with quantitative trait loci for ear wound-healing. Ectopic treatment with one of these proteins, complement factor H (CFH), accelerated wound repair and induced regeneration in typically fibrotic wounds. Through single-cell RNA sequencing (RNA-seq), we observed that CFH treatment dramatically reduced immune cell recruitment to wounds, suggesting a potential mechanism for CFH's effect. Overall, our results provide insights into the molecular drivers of regeneration with potential clinical implications.
Collapse
Affiliation(s)
- Katya L Mack
- Stanford University, Department of Biology, Stanford, CA, USA
| | - Heather E Talbott
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA; Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA
| | - Michelle F Griffin
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Jennifer B L Parker
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA; Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA
| | - Nicholas J Guardino
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Amanda F Spielman
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Michael F Davitt
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Shamik Mascharak
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA; Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA
| | - Mauricio Downer
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Annah Morgan
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Caleb Valencia
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Deena Akras
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Mark J Berger
- Stanford University, Department of Computer Science, Stanford, CA 94305, USA
| | - Derrick C Wan
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA
| | - Hunter B Fraser
- Stanford University, Department of Biology, Stanford, CA, USA.
| | - Michael T Longaker
- Stanford School of Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford, CA, USA; Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA.
| |
Collapse
|
4
|
Thorup AS, Dell'Accio F, Eldridge SE. A Mouse Model of Acute Cartilage Injury and Repair. Methods Mol Biol 2023; 2598:337-344. [PMID: 36355303 DOI: 10.1007/978-1-0716-2839-3_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Chondral defects are common and disabling. The development of pharmacological approaches for cartilage repair requires the availability of in vivo models which are amenable for gain and loss of function and ideally to genetic modification. In this chapter, we describe a method to induce full-thickness cartilage defects which, in young DBA/1 mice, heal spontaneously, but fail to heal in C57BL/6 mice of the same age or in aged DBA/1 mice. This model (or variants) has been used for genetic screenings to identify genes associated to repair capacity, to study stem cells involved in cartilage repair, and to study the function of molecules involved in repair mechanisms.
Collapse
Affiliation(s)
- Anne-Sophie Thorup
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Francesco Dell'Accio
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | - Suzanne E Eldridge
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
5
|
Zebrowitz E, Aslanukov A, Kajikawa T, Bedelbaeva K, Bollinger S, Zhang Y, Sarfatti D, Cheng J, Messersmith PB, Hajishengallis G, Heber-Katz E. Prolyl-hydroxylase inhibitor-induced regeneration of alveolar bone and soft tissue in a mouse model of periodontitis through metabolic reprogramming. FRONTIERS IN DENTAL MEDICINE 2022; 3:992722. [PMID: 37641630 PMCID: PMC10462383 DOI: 10.3389/fdmed.2022.992722] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023] Open
Abstract
Bone injuries and fractures reliably heal through a process of regeneration with restoration to original structure and function when the gap between adjacent sides of a fracture site is small. However, when there is significant volumetric loss of bone, bone regeneration usually does not occur. In the present studies, we explore a particular case of volumetric bone loss in a mouse model of human periodontal disease (PD) in which alveolar bone surrounding teeth is permanently lost and not replaced. This model employs the placement a ligature around the upper second molar for 10 days leading to inflammation and bone breakdown and faithfully replicates the bacterially-induced inflammatory etiology of human PD to induce bone degeneration. After ligature removal, mice are treated with a timed-release formulation of a small molecule inhibitor of prolylhydroxylases (PHDi; 1,4-DPCA) previously shown to induce epimorphic regeneration of soft tissue in non-regenerating mice. This PHDi induces high expression of HIF-1α and is able to shift the metabolic state from OXPHOS to aerobic glycolysis, an energetic state used by stem cells and embryonic tissue. This regenerative response was completely blocked by siHIF1a. In these studies, we show that timed-release 1,4-DPCA rapidly and completely restores PD-affected bone and soft tissue with normal anatomic fidelity and with increased stem cell markers due to site-specific stem cell migration and/or de-differentiation of local tissue, periodontal ligament (PDL) cell proliferation, and increased vascularization. In-vitro studies using gingival tissue show that 1,4-DPCA indeed induces de-differentiation and the expression of stem cell markers but does not exclude the role of migrating stem cells. Evidence of metabolic reprogramming is seen by the expression of not only HIF-1a, its gene targets, and resultant de-differentiation markers, but also the metabolic genes Glut-1, Gapdh, Pdk1, Pgk1 and Ldh-a in jaw periodontal tissue.
Collapse
Affiliation(s)
- Elan Zebrowitz
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania, United States of America
- Current address: New York Medical College, 40 Sunshine Cottage Rd, Valhalla New York, United States of America
| | - Azamat Aslanukov
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania, United States of America
| | - Tetsuhiro Kajikawa
- University of Pennsylvania School of Dental Medicine, Department of Basic and Translational Sciences, Philadelphia, Pennsylvania, United States of America
| | - Kamila Bedelbaeva
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania, United States of America
| | - Sam Bollinger
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania, United States of America
- Current address: Cancer Biology Graduate Group, Stanford, California, United States of America
| | - Yong Zhang
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania, United States of America
- Current address: Rockland Immunochemicals, Inc., Limerick, Pennsylvania, United States of America
| | - David Sarfatti
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania, United States of America
| | - Jing Cheng
- Materials Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
- Current address: Alcon Laboratories, 11460 Johns Creek Pkwy, Duluth, Georgia, United States of America
| | - Phillip B. Messersmith
- Department of Bioengineering and Materials Science and Engineering, UC Berkeley, Berkeley California, United States of America
- Materials Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - George Hajishengallis
- University of Pennsylvania School of Dental Medicine, Department of Basic and Translational Sciences, Philadelphia, Pennsylvania, United States of America
| | - Ellen Heber-Katz
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania, United States of America
| |
Collapse
|
6
|
Mascharak S, desJardins-Park HE, Davitt MF, Guardino NJ, Gurtner GC, Wan DC, Longaker MT. Modulating Cellular Responses to Mechanical Forces to Promote Wound Regeneration. Adv Wound Care (New Rochelle) 2022; 11:479-495. [PMID: 34465219 PMCID: PMC9245727 DOI: 10.1089/wound.2021.0040] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 08/23/2021] [Indexed: 12/13/2022] Open
Abstract
Significance: Skin scarring poses a major biomedical burden for hundreds of millions of patients annually. However, this burden could be mitigated by therapies that promote wound regeneration, with full recovery of skin's normal adnexa, matrix ultrastructure, and mechanical strength. Recent Advances: The observation of wound regeneration in several mouse models suggests a retained capacity for postnatal mammalian skin to regenerate under the right conditions. Mechanical forces are a major contributor to skin fibrosis and a prime target for devices and therapeutics that could promote skin regeneration. Critical Issues: Wound-induced hair neogenesis, Acomys "spiny" mice, Murphy Roths Large mice, and mice treated with mechanotransduction inhibitors all show various degrees of wound regeneration. Comparison of regenerating wounds in these models against scarring wounds reveals differences in extracellular matrix interactions and in mechanosensitive activation of key signaling pathways, including Wnt, Sonic hedgehog, focal adhesion kinase, and Yes-associated protein. The advent of single-cell "omics" technologies has deepened this understanding and revealed that regeneration may recapitulate development in certain contexts, although it is unknown whether these mechanisms are relevant to healing in tight-skinned animals such as humans. Future Directions: While early findings in mice are promising, comparison across model systems is needed to resolve conflicting mechanisms and to identify conserved master regulators of skin regeneration. There also remains a dire need for studies on mechanomodulation of wounds in large, tight-skinned animals, such as red Duroc pigs, which better approximate human wound healing.
Collapse
Affiliation(s)
- Shamik Mascharak
- Division of Plastic and Reconstructive Surgery, Department of Surgery; Stanford, California, USA
- Institute for Stem Cell Biology and Regenerative Medicine; Stanford University School of Medicine, Stanford, California, USA
| | - Heather E. desJardins-Park
- Division of Plastic and Reconstructive Surgery, Department of Surgery; Stanford, California, USA
- Institute for Stem Cell Biology and Regenerative Medicine; Stanford University School of Medicine, Stanford, California, USA
| | - Michael F. Davitt
- Division of Plastic and Reconstructive Surgery, Department of Surgery; Stanford, California, USA
| | - Nicholas J. Guardino
- Division of Plastic and Reconstructive Surgery, Department of Surgery; Stanford, California, USA
| | - Geoffrey C. Gurtner
- Division of Plastic and Reconstructive Surgery, Department of Surgery; Stanford, California, USA
| | - Derrick C. Wan
- Division of Plastic and Reconstructive Surgery, Department of Surgery; Stanford, California, USA
| | - Michael T. Longaker
- Division of Plastic and Reconstructive Surgery, Department of Surgery; Stanford, California, USA
- Institute for Stem Cell Biology and Regenerative Medicine; Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
7
|
Porto A, Rolfe S, Maga AM. ALPACA: A fast and accurate computer vision approach for automated landmarking of three-dimensional biological structures. Methods Ecol Evol 2021; 12:2129-2144. [PMID: 35874971 PMCID: PMC9291522 DOI: 10.1111/2041-210x.13689] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2021] [Indexed: 11/27/2022]
Abstract
Landmark-based geometric morphometrics has emerged as an essential discipline for the quantitative analysis of size and shape in ecology and evolution. With the ever-increasing density of digitized landmarks, the possible development of a fully automated method of landmark placement has attracted considerable attention. Despite the recent progress in image registration techniques, which could provide a pathway to automation, three-dimensional (3D) morphometric data are still mainly gathered by trained experts. For the most part, the large infrastructure requirements necessary to perform image-based registration, together with its system specificity and its overall speed, have prevented its wide dissemination.Here, we propose and implement a general and lightweight point cloud-based approach to automatically collect high-dimensional landmark data in 3D surfaces (Automated Landmarking through Point cloud Alignment and Correspondence Analysis). Our framework possesses several advantages compared with image-based approaches. First, it presents comparable landmarking accuracy, despite relying on a single, random reference specimen and much sparser sampling of the structure's surface. Second, it can be efficiently run on consumer-grade personal computers. Finally, it is general and can be applied at the intraspecific level to any biological structure of interest, regardless of whether anatomical atlases are available.Our validation procedures indicate that the method can recover intraspecific patterns of morphological variation that are largely comparable to those obtained by manual digitization, indicating that the use of an automated landmarking approach should not result in different conclusions regarding the nature of multivariate patterns of morphological variation.The proposed point cloud-based approach has the potential to increase the scale and reproducibility of morphometrics research. To allow ALPACA to be used out-of-the-box by users with no prior programming experience, we implemented it as a SlicerMorph module. SlicerMorph is an extension that enables geometric morphometrics data collection and 3D specimen analysis within the open-source 3D Slicer biomedical visualization ecosystem. We expect that convenient access to this platform will make ALPACA broadly applicable within ecology and evolution.
Collapse
Affiliation(s)
- Arthur Porto
- Department of Biological SciencesLouisiana State UniversityBaton RougeLAUSA
- Center for Computation and TechnologyLouisiana State UniversityBaton RougeLAUSA
| | - Sara Rolfe
- Friday Harbor LaboratoriesUniversity of WashingtonSan Juan IslandWAUSA
- Center for Development Biology and Regenerative MedicineSeattle Children's Research InstituteSeattleWAUSA
| | - A. Murat Maga
- Center for Development Biology and Regenerative MedicineSeattle Children's Research InstituteSeattleWAUSA
- Division of Craniofacial MedicineDepartment of PediatricsUniversity of WashingtonSeattleWAUSA
| |
Collapse
|
8
|
Thomas BL, Eldridge SE, Nosrati B, Alvarez M, Thorup A, Nalesso G, Caxaria S, Barawi A, Nicholson JG, Perretti M, Gaston‐Massuet C, Pitzalis C, Maloney A, Moore A, Jupp R, Dell'Accio F. WNT3A-loaded exosomes enable cartilage repair. J Extracell Vesicles 2021; 10:e12088. [PMID: 34025953 PMCID: PMC8134720 DOI: 10.1002/jev2.12088] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 09/06/2020] [Accepted: 09/08/2020] [Indexed: 12/13/2022] Open
Abstract
Cartilage defects repair poorly. Recent genetic studies suggest that WNT3a may contribute to cartilage regeneration, however the dense, avascular cartilage extracellular matrix limits its penetration and signalling to chondrocytes. Extracellular vesicles actively penetrate intact cartilage. This study investigates the effect of delivering WNT3a into large cartilage defects in vivo using exosomes as a delivery vehicle. Exosomes were purified by ultracentrifugation from conditioned medium of either L-cells overexpressing WNT3a or control un-transduced L-cells, and characterized by electron microscopy, nanoparticle tracking analysis and marker profiling. WNT3a loaded on exosomes was quantified by western blotting and functionally characterized in vitro using the SUPER8TOPFlash reporter assay and other established readouts including proliferation and proteoglycan content. In vivo pathway activation was assessed using TCF/Lef:H2B-GFP reporter mice. Wnt3a loaded exosomes were injected into the knees of mice, in which large osteochondral defects were surgically generated. The degree of repair was histologically scored after 8 weeks. WNT3a was successfully loaded on exosomes and resulted in activation of WNT signalling in vitro. In vivo, recombinant WNT3a failed to activate WNT signalling in cartilage, whereas a single administration of WNT3a loaded exosomes activated canonical WNT signalling for at least one week, and eight weeks later, improved the repair of osteochondral defects. WNT3a assembled on exosomes, is efficiently delivered into cartilage and contributes to the healing of osteochondral defects.
Collapse
Affiliation(s)
- Bethan L. Thomas
- Barts and the London School of Medicine and DentistryWilliam Harvey Research InstituteQueen Mary University of LondonLondonUK
| | - Suzanne E. Eldridge
- Barts and the London School of Medicine and DentistryWilliam Harvey Research InstituteQueen Mary University of LondonLondonUK
| | - Babak Nosrati
- Dipartimento di scienza e tecnologia del farmacoUniversità degli Studi di TorinoTorinoItaly
| | - Mario Alvarez
- Barts and the London School of Medicine and DentistryWilliam Harvey Research InstituteQueen Mary University of LondonLondonUK
| | - Anne‐Sophie Thorup
- Barts and the London School of Medicine and DentistryWilliam Harvey Research InstituteQueen Mary University of LondonLondonUK
| | - Giovanna Nalesso
- School of Veterinary MedicineUniversity of SurreyDaphne Jackson RoadGuildfordUK
| | - Sara Caxaria
- Barts and the London School of Medicine and DentistryWilliam Harvey Research InstituteQueen Mary University of LondonLondonUK
| | - Aida Barawi
- Barts and the London School of Medicine and DentistryWilliam Harvey Research InstituteQueen Mary University of LondonLondonUK
| | - James G. Nicholson
- Barts and the London School of Medicine and DentistryWilliam Harvey Research InstituteQueen Mary University of LondonLondonUK
| | - Mauro Perretti
- Barts and the London School of Medicine and DentistryWilliam Harvey Research InstituteQueen Mary University of LondonLondonUK
| | - Carles Gaston‐Massuet
- Barts and the London School of Medicine and DentistryWilliam Harvey Research InstituteQueen Mary University of LondonLondonUK
| | - Costantino Pitzalis
- Barts and the London School of Medicine and DentistryWilliam Harvey Research InstituteQueen Mary University of LondonLondonUK
| | | | | | | | - Francesco Dell'Accio
- Barts and the London School of Medicine and DentistryWilliam Harvey Research InstituteQueen Mary University of LondonLondonUK
| |
Collapse
|
9
|
Rai MF, Cheverud JM, Schmidt EJ, Sandell LJ. Genetic correlations between cartilage regeneration and degeneration reveal an inverse relationship. Osteoarthritis Cartilage 2020; 28:1111-1120. [PMID: 32437968 PMCID: PMC7387169 DOI: 10.1016/j.joca.2020.04.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 04/24/2020] [Accepted: 04/29/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The etiology of osteoarthritis (OA) is unknown, however, there appears to be a significant contribution from genetics. We have identified recombinant inbred strains of mice derived from LG/J (large) and SM/J (small) strains that vary significantly in their ability to repair articular cartilage and susceptibility to post-traumatic OA due to their genetic composition. Here, we report cartilage repair phenotypes in the same strains of mice in which OA susceptibility was analyzed previously, and determine the genetic correlations between phenotypes. DESIGN We used 12 recombinant inbred strains, including the parental strains, to test three phenotypes: ear-wound healing (n = 263), knee articular cartilage repair (n = 131), and post-traumatic OA (n = 53) induced by the surgical destabilization of the medial meniscus (DMM). Genetic correlations between various traits were calculated as Pearson's correlation coefficients of strain means. RESULTS We found a significant positive correlation between ear-wound healing and articular cartilage regeneration (r = 0.71; P = 0.005). We observed a strong inverse correlation between articular cartilage regeneration and susceptibility to OA based on maximum (r = -0.54; P = 0.036) and summed Osteoarthritis Research Society International (OARSI) scores (r = -0.56; P = 0.028). Synovitis was not significantly correlated with articular cartilage regeneration but was significantly positively correlated with maximum (r = 0.63; P = 0.014) and summed (r = 0.70; P = 0.005) OARSI scores. Ectopic calcification was significantly positively correlated with articular cartilage regeneration (r = 0.59; P = 0.021). CONCLUSIONS Using recombinant inbred strains, our study allows, for the first time, the measurement of genetic correlations of regeneration phenotypes with degeneration phenotypes, characteristic of OA (cartilage degeneration, synovitis). We demonstrate that OA is positively correlated with synovitis and inversely correlated with the ability to repair cartilage. These results suggest an addition to the risk paradigm for OA from a focus on degeneration to regeneration.
Collapse
Affiliation(s)
- Muhammad Farooq Rai
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, United States,Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, United States
| | - James M. Cheverud
- Department of Biology, Loyola University, Chicago, IL, United States
| | - Eric J. Schmidt
- School of Physician Assistant Medicine, College of Health Sciences, University of Lynchburg, Lynchburg, VA, United States
| | - Linda J. Sandell
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
10
|
Duan X, Cai L, Schmidt EJ, Shen J, Tycksen ED, O’Keefe R, Cheverud JM, Farooq Rai M. RNA-seq analysis of chondrocyte transcriptome reveals genetic heterogeneity in LG/J and SM/J murine strains. Osteoarthritis Cartilage 2020; 28:516-527. [PMID: 31945456 PMCID: PMC7108965 DOI: 10.1016/j.joca.2020.01.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/02/2020] [Accepted: 01/03/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To investigate the transcriptomic differences in chondrocytes obtained from LG/J (large, healer) and SM/J (small, non-healer) murine strains in an attempt to discern the molecular pathways implicated in cartilage regeneration and susceptibility to osteoarthritis (OA). DESIGN We performed RNA-sequencing on chondrocytes derived from LG/J (n = 16) and SM/J (n = 16) mice. We validated the expression of candidate genes and compared single nucleotide polymorphisms (SNPs) between the two mouse strains. We also examined gene expression of positional candidates for ear pinna regeneration and long bone length quantitative trait loci (QTLs) that display differences in cartilaginous expression. RESULTS We observed a distinct genetic heterogeneity between cells derived from LG/J and SM/J mouse strains. We found that gene ontologies representing cell development, cartilage condensation, and regulation of cell differentiation were enriched in LG/J chondrocytes. In contrast, gene ontologies enriched in the SM/J chondrocytes were mainly related to inflammation and degeneration. Moreover, SNP analysis revealed that multiple validated genes vary in sequence between LG/J and SM/J in coding and highly conserved noncoding regions. Finally, we showed that most QTLs have 20-30% of their positional candidates displaying differential expression between the two mouse strains. CONCLUSIONS While the enrichment of pathways related to cell differentiation, cartilage development and cartilage condensation infers superior healing potential of LG/J strain, the enrichment of pathways related to cytokine production, immune cell activation and inflammation entails greater susceptibility of SM/J strain to OA. These data provide novel insights into chondrocyte transcriptome and aid in identification of the quantitative trait genes and molecular differences underlying the phenotypic differences associated with individual QTLs.
Collapse
Affiliation(s)
- Xin Duan
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Lei Cai
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Eric J. Schmidt
- School of Physician Assistant Medicine, College of Health Sciences, University of Lynchburg, Lynchburg, VA, United States
| | - Jie Shen
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Eric D. Tycksen
- Genome Technology Access Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Regis O’Keefe
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, United States
| | - James M. Cheverud
- Department of Biology, Loyola University, Chicago, IL, United States
| | - Muhammad Farooq Rai
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, United States, Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
11
|
Abstract
In this chapter we will review both the rationale and experimental design for using Heterogeneous Stock (HS) populations for fine-mapping of complex traits in mice and rats. We define an HS as an outbred population derived from an intercross between two or more inbred strains. HS have been used to perform genome-wide association studies (GWAS) for multiple behavioral, physiological, and gene expression traits. GWAS using HS require four key steps, which we review: selection of an appropriate HS population, phenotyping, genotyping, and statistical analysis. We provide advice on the selection of an HS, comment on key issues related to phenotyping, discuss genotyping methods relevant to these populations, and describe statistical genetic analyses that are applicable to genetic analyses that use HS.
Collapse
|
12
|
Silva MJ, Eekhoff JD, Patel T, Kenney-Hunt JP, Brodt MD, Steger-May K, Scheller EL, Cheverud JM. Effects of High-Fat Diet and Body Mass on Bone Morphology and Mechanical Properties in 1100 Advanced Intercross Mice. J Bone Miner Res 2019; 34:711-725. [PMID: 30615803 PMCID: PMC6879418 DOI: 10.1002/jbmr.3648] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/09/2018] [Accepted: 11/19/2018] [Indexed: 01/19/2023]
Abstract
Obesity is generally protective against osteoporosis and bone fracture. However, recent studies indicate that the influence of obesity on the skeleton is complex and can be detrimental. We evaluated the effects of a high-fat, obesogenic diet on the femur and radius of 1100 mice (males and females) from the Large-by-Small advanced intercross line (F34 generation). At age 5 months, bone morphology was assessed by microCT and mechanical properties by three-point bending. Mice raised on a high-fat diet had modestly greater cortical area, bending stiffness, and strength. Size-independent material properties were unaffected by a high-fat diet, indicating that diet influenced bone quantity but not quality. Bone size and mechanical properties were strongly correlated with body mass. However, the increases in many bone traits per unit increase in body mass were less in high-fat diet mice than low-fat diet mice. Thus, although mice raised on a high-fat diet have, on average, bigger and stronger bones than low-fat-fed mice, a high-fat diet diminished the positive relationship between body mass and bone size and whole-bone strength. The findings support the concept that there are diminishing benefits to skeletal health with increasing obesity. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Matthew J Silva
- Department of Orthopaedic Surgery, Washington University in Saint Louis, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO, USA
| | - Jeremy D Eekhoff
- Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO, USA
| | - Tarpit Patel
- Department of Orthopaedic Surgery, Washington University in Saint Louis, St. Louis, MO, USA
| | - Jane P Kenney-Hunt
- Department of Anatomy and Neurobiology, Washington University in Saint Louis, St. Louis, MO, USA
| | - Michael D Brodt
- Department of Orthopaedic Surgery, Washington University in Saint Louis, St. Louis, MO, USA
| | - Karen Steger-May
- Division of Biostatistics, Washington University in Saint Louis, St. Louis, MO, USA
| | - Erica L Scheller
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University in Saint Louis, St. Louis, MO, USA
| | - James M Cheverud
- Department of Anatomy and Neurobiology, Washington University in Saint Louis, St. Louis, MO, USA.,Department of Biology, Loyola University Chicago, Chicago, IL, USA
| |
Collapse
|
13
|
Bagnati M, Moreno-Moral A, Ko JH, Nicod J, Harmston N, Imprialou M, Game L, Gil J, Petretto E, Behmoaras J. Systems genetics identifies a macrophage cholesterol network associated with physiological wound healing. JCI Insight 2019; 4:e125736. [PMID: 30674726 PMCID: PMC6413785 DOI: 10.1172/jci.insight.125736] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 12/18/2018] [Indexed: 01/18/2023] Open
Abstract
Among other cells, macrophages regulate the inflammatory and reparative phases during wound healing but genetic determinants and detailed molecular pathways that modulate these processes are not fully elucidated. Here, we took advantage of normal variation in wound healing in 1,378 genetically outbred mice, and carried out macrophage RNA-sequencing profiling of mice with extreme wound healing phenotypes (i.e., slow and fast healers, n = 146 in total). The resulting macrophage coexpression networks were genetically mapped and led to the identification of a unique module under strong trans-acting genetic control by the Runx2 locus. This macrophage-mediated healing network was specifically enriched for cholesterol and fatty acid biosynthetic processes. Pharmacological blockage of fatty acid synthesis with cerulenin resulted in delayed wound healing in vivo, and increased macrophage infiltration in the wounded skin, suggesting the persistence of an unresolved inflammation. We show how naturally occurring sequence variation controls transcriptional networks in macrophages, which in turn regulate specific metabolic pathways that could be targeted in wound healing.
Collapse
Affiliation(s)
- Marta Bagnati
- Centre for Inflammatory Disease, Imperial College London, Hammersmith Hospital, London, United Kingdom (UK)
| | | | - Jeong-Hun Ko
- Centre for Inflammatory Disease, Imperial College London, Hammersmith Hospital, London, United Kingdom (UK)
| | - Jérôme Nicod
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | | | - Martha Imprialou
- Centre for Inflammatory Disease, Imperial College London, Hammersmith Hospital, London, United Kingdom (UK)
| | - Laurence Game
- Genomics Laboratory, Medical Research Council (MRC) London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Jesus Gil
- Cell Proliferation Group, MRC London Institute of Medical Sciences (LMS), London, UK
| | - Enrico Petretto
- Duke-NUS Medical School, Singapore, Singapore
- MRC London Institute of Medical Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Jacques Behmoaras
- Centre for Inflammatory Disease, Imperial College London, Hammersmith Hospital, London, United Kingdom (UK)
| |
Collapse
|
14
|
Heber-Katz E, Messersmith P. Drug delivery and epimorphic salamander-type mouse regeneration: A full parts and labor plan. Adv Drug Deliv Rev 2018. [PMID: 29524586 DOI: 10.1016/j.addr.2018.02.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The capacity to regenerate entire body parts, tissues, and organs had generally been thought to be lost in evolution with very few exceptions (e.g. the liver) surviving in mammals. The discovery of the MRL mouse and the elucidation of the underlying molecular pathway centering around hypoxia inducible factor, HIF-1α, has allowed a drug and materials approach to regeneration in mice and hopefully humans. The HIF-1α pathway is ancient and permitted the transition from unicellular to multicellular organisms. Furthermore, HIF-1α and its regulation by PHDs, important oxygen sensors in the cell, provides a perfect drug target. We review the historical background of regeneration biology, the discovery of the MRL mouse, and its underlying biology, and novel approaches to drugs, targets, and delivery systems (see Fig. 1).
Collapse
|
15
|
Heber-Katz E. Oxygen, Metabolism, and Regeneration: Lessons from Mice. Trends Mol Med 2017; 23:1024-1036. [PMID: 28988849 DOI: 10.1016/j.molmed.2017.08.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 08/05/2017] [Accepted: 08/20/2017] [Indexed: 12/12/2022]
Abstract
The discovery that the Murphy Roths Large (MRL) mouse strain is a fully competent, epimorphic tissue regenerator, proved that the machinery of regeneration was preserved through evolution from hydra, to salamanders, to mammals. Such concepts have allowed translation of the biology of amphibians, and their ability to regenerate, to a mammalian context. We identified the ancient hypoxia-inducible factor (HIF)-1α pathway, operating through prolyl hydroxylase domain proteins (PHDs), as a central player in mouse regeneration. Thus, the possibility of targeting PHDs or other HIF-1α modifiers to effectively recreate the amphibian regenerative state has emerged. We posit that these regenerative pathways are critical in mammals. Moreover, the current approved use of PHD inhibitors in the clinic should allow fast-track translation from mouse studies to drug-based regenerative therapy in humans.
Collapse
Affiliation(s)
- Ellen Heber-Katz
- Laboratory of Regenerative Medicine, Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA.
| |
Collapse
|
16
|
Godwin JW, Pinto AR, Rosenthal NA. Chasing the recipe for a pro-regenerative immune system. Semin Cell Dev Biol 2016; 61:71-79. [PMID: 27521522 DOI: 10.1016/j.semcdb.2016.08.008] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 08/08/2016] [Accepted: 08/09/2016] [Indexed: 02/07/2023]
Abstract
Identification of the key ingredients and essential processes required to achieve perfect tissue regeneration in humans has so far remained elusive. Injury in vertebrates induces an obligatory wound response that will precede or overlap any regeneration specific program or scarring outcome. This process shapes the cellular and molecular landscape of the tissue, influencing the success of endogenous repair pathways or for potential clinical intervention. The involvement of immune cells is also required for aspects of development extending beyond the initial inflammatory phase of wounding. It has now become clear from amphibian, fish and mammalian models of tissue injury that the type of immune response and the profile of immune cells attending the site of injury can act as the gatekeepers that determine wound repair quality. The heterogeneity among innate and adaptive immune cell populations, along with the developmental origin of these cells, form key ingredients affecting the potential for downstream repair and the suppression of fibrosis. Cell-to-cell interactions between immune cells, such as macrophages and T cells, with stem cells and mesenchymal cells are critically important for shaping this process and these exchanges, are in turn influenced by the type of injury, tissue location and developmental stage of the organism. Developmentally, mouse cardiac regeneration is restricted to early stages of postnatal life where the balance of innate to adaptive immune cells may be poised towards regeneration. In the injured adult mouse liver, specific macrophage subsets improve repair while other bone marrow derived cells can exacerbate injury. Other studies using genetically diverse mice have shown enhanced regeneration in certain strains, restricted to specific tissues. This enhanced repair is linked with expression of genes such as Insulin-like Growth Factor- 1 (IGF-1) and activin (Act 1), that both play important roles in shaping the immune system. Immune cells are now appreciated to have powerful influences on critical cell types required for regeneration success. The winning recipe for tissue regeneration is likely to be found ultimately by identifying the genetic elements and specific cell populations that limit or allow intrinsic potential. This will be essential for developing therapeutic strategies for tissue regeneration in humans.
Collapse
Affiliation(s)
- James W Godwin
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA; MDI Biological Laboratory, Salisbury Cove, ME 04672, USA; Australian Regenerative Medicine Institute, Monash University, Victoria, 3800, Australia.
| | | | | |
Collapse
|
17
|
Enhanced cartilage repair in 'healer' mice-New leads in the search for better clinical options for cartilage repair. Semin Cell Dev Biol 2016; 62:78-85. [PMID: 27130635 DOI: 10.1016/j.semcdb.2016.04.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 04/25/2016] [Indexed: 12/13/2022]
Abstract
Adult articular cartilage has a poor capacity to undergo intrinsic repair. Current strategies for the repair of large cartilage defects are generally unsatisfactory because the restored cartilage does not have the same resistance to biomechanical loading as authentic articular cartilage and degrades over time. Recently, an exciting new research direction, focused on intrinsic cartilage regeneration rather than fibrous repair by external means, has emerged. This review explores the new findings in this rapidly moving field as they relate to the clinical goal of restoration of structurally robust, stable and non-fibrous articular cartilage following injury.
Collapse
|
18
|
Zhang Y, Strehin I, Bedelbaeva K, Gourevitch D, Clark L, Leferovich J, Messersmith PB, Heber-Katz E. Drug-induced regeneration in adult mice. Sci Transl Med 2016; 7:290ra92. [PMID: 26041709 DOI: 10.1126/scitranslmed.3010228] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Whereas amphibians regenerate lost appendages spontaneously, mammals generally form scars over the injury site through the process of wound repair. The MRL mouse strain is an exception among mammals because it shows a spontaneous regenerative healing trait and so can be used to investigate proregenerative interventions in mammals. We report that hypoxia-inducible factor 1α (HIF-1α) is a central molecule in the process of regeneration in adult MRL mice. The degradation of HIF-1α protein, which occurs under normoxic conditions, is mediated by prolyl hydroxylases (PHDs). We used the drug 1,4-dihydrophenonthrolin-4-one-3-carboxylic acid (1,4-DPCA), a PHD inhibitor, to stabilize constitutive expression of HIF-1α protein. A locally injectable hydrogel containing 1,4-DPCA was designed to achieve controlled delivery of the drug over 4 to 10 days. Subcutaneous injection of the 1,4-DPCA/hydrogel into Swiss Webster mice that do not show a regenerative phenotype increased stable expression of HIF-1α protein over 5 days, providing a functional measure of drug release in vivo. Multiple peripheral subcutaneous injections of the 1,4-DPCA/hydrogel over a 10-day period led to regenerative wound healing in Swiss Webster mice after ear hole punch injury. Increased expression of the HIF-1α protein may provide a starting point for future studies on regeneration in mammals.
Collapse
Affiliation(s)
- Yong Zhang
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Iossif Strehin
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Khamilia Bedelbaeva
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Dmitri Gourevitch
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Lise Clark
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - John Leferovich
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Phillip B Messersmith
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Ellen Heber-Katz
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA 19104, USA.
| |
Collapse
|
19
|
Rai MF, Schmidt EJ, Hashimoto S, Cheverud JM, Sandell LJ. Genetic loci that regulate ectopic calcification in response to knee trauma in LG/J by SM/J advanced intercross mice. J Orthop Res 2015; 33:1412-23. [PMID: 25989359 PMCID: PMC5025301 DOI: 10.1002/jor.22944] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 05/10/2015] [Indexed: 02/04/2023]
Abstract
This study reports on genetic susceptibility to ectopic calcification in the LG/J and SM/J advanced intercross mice. Using 347 mice in 98 full-sibships, destabilization of medial meniscus (DMM) was performed to induce joint injury. We found that joint destabilization instigated ectopic calcifications as detected and quantified by micro-CT. We performed quantitative trait locus (QTL) analysis to map ectopic calcification phenotypes to discrete genomic locations. To validate the functional significance of the selected QTL candidate genes, we compared mRNA expression between parental LG/J and SM/J inbred strains. Overall, we detected 20 QTLs affecting synovial and meniscus calcification phenotypes with 11 QTLs linked to synovial calcification. Functional and bioinformatic analyses of single nucleotide polymorphism (SNP) identified functional classifications relevant to angiogenesis (Myo1e, Kif26b, Nprl3, Stab2, Fam105b), bone metabolism/calcification (Tle3, Tgfb2, Lipc, Nfe2l1, Ank, Fam105b), arthritis (Stab2, Tbx21, Map4k4, Hoxb9, Larp6, Col1a2, Adam10, Timp3, Nfe2l1, Trpm3), and ankylosing-spondylitis (Ank, Pon1, Il1r2, Tbkbp1) indicating that ectopic calcification involves multiple mechanisms. Furthermore, the expression of 11 out of 78 candidate genes was significantly different between LG/J and SM/J. Correlation analysis showed that Aff3, Fam81a, Syn3, and Ank were correlated with synovial calcification. Taken together, our findings of multiple genetic loci suggest the involvement of multiple genes contributing to ectopic calcification.
Collapse
Affiliation(s)
- Muhammad Farooq Rai
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine at Barnes-Jewish Hospital, 425 S. Euclid Avenue MS 8233, St. Louis, MO 63110, USA
| | - Eric J. Schmidt
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine at Barnes-Jewish Hospital, 425 S. Euclid Avenue MS 8233, St. Louis, MO 63110, USA
| | - Shingo Hashimoto
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine at Barnes-Jewish Hospital, 425 S. Euclid Avenue MS 8233, St. Louis, MO 63110, USA
| | - James M. Cheverud
- Department of Biology, Loyola University, 1050 W. Sheridan Road, Chicago, IL 60660, USA
| | - Linda J. Sandell
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine at Barnes-Jewish Hospital, 425 S. Euclid Avenue MS 8233, St. Louis, MO 63110, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine at Barnes-Jewish Hospital, 425 S. Euclid Avenue MS 8233, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University School of Medicine at Barnes-Jewish Hospital, 425 S. Euclid Avenue MS 8233, St. Louis, MO 63110, USA
| |
Collapse
|
20
|
Heber-Katz E. From Immunity and Vaccines to Mammalian Regeneration. J Infect Dis 2015; 212 Suppl 1:S52-8. [PMID: 26116734 DOI: 10.1093/infdis/jiu637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Our current understanding of major histocompatibility complex (MHC)-mediated antigen presentation in self and nonself immune recognition was derived from immunological studies of autoimmunity and virus-host interactions, respectively. The trimolecular complex of the MHC molecule, antigen, and T-cell receptor accounts for the phenomena of immunodominance and MHC degeneracy in both types of responses and constrains vaccine development. Out of such considerations, we developed a simple peptide vaccine construct that obviates immunodominance, resulting in a broadly protective T-cell response in the absence of antibody. In the course of autoimmunity studies, we identified the MRL mouse strain as a mammalian model of amphibian-like regeneration. A significant level of DNA damage in the cells from this mouse pointed to the role of the cell cycle checkpoint gene CDKN1a, or p21(cip1/waf1). The MRL mouse has highly reduced levels of this molecule, and a genetic knockout of this single gene in otherwise nonregenerating strains led to an MRL-type regenerative response, indicating that the ability to regenerate has not been lost during evolution.
Collapse
|
21
|
Nikolskiy I, Conrad DF, Chun S, Fay JC, Cheverud JM, Lawson HA. Using whole-genome sequences of the LG/J and SM/J inbred mouse strains to prioritize quantitative trait genes and nucleotides. BMC Genomics 2015; 16:415. [PMID: 26016481 PMCID: PMC4445795 DOI: 10.1186/s12864-015-1592-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 04/28/2015] [Indexed: 12/04/2022] Open
Abstract
Background The laboratory mouse is the most commonly used model for studying variation in complex traits relevant to human disease. Here we present the whole-genome sequences of two inbred strains, LG/J and SM/J, which are frequently used to study variation in complex traits as diverse as aging, bone-growth, adiposity, maternal behavior, and methamphetamine sensitivity. Results We identified small nucleotide variants (SNVs) and structural variants (SVs) in the LG/J and SM/J strains relative to the reference genome and discovered novel variants in these two strains by comparing their sequences to other mouse genomes. We find that 39% of the LG/J and SM/J genomes are identical-by-descent (IBD). We characterized amino-acid changing mutations using three algorithms: LRT, PolyPhen-2 and SIFT. We also identified polymorphisms between LG/J and SM/J that fall in regulatory regions and highly informative transcription factor binding sites (TFBS). We intersected these functional predictions with quantitative trait loci (QTL) mapped in advanced intercrosses of these two strains. We find that QTL are both over-represented in non-IBD regions and highly enriched for variants predicted to have a functional impact. Variants in QTL associated with metabolic (231 QTL identified in an F16 generation) and developmental (41 QTL identified in an F34 generation) traits were interrogated and we highlight candidate quantitative trait genes (QTG) and nucleotides (QTN) in a QTL on chr13 associated with variation in basal glucose levels and in a QTL on chr6 associated with variation in tibia length. Conclusions We show how integrating genomic sequence with QTL reduces the QTL search space and helps researchers prioritize candidate genes and nucleotides for experimental follow-up. Additionally, given the LG/J and SM/J phylogenetic context among inbred strains, these data contribute important information to the genomic landscape of the laboratory mouse. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1592-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Igor Nikolskiy
- Department of Genetics, Washington University School of Medicine, Campus Box 8108, 660 S Euclid Ave, St Louis, MO, 63110, USA.
| | - Donald F Conrad
- Department of Genetics, Washington University School of Medicine, Campus Box 8108, 660 S Euclid Ave, St Louis, MO, 63110, USA.
| | - Sung Chun
- Division of Genetics, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Justin C Fay
- Department of Genetics, Washington University School of Medicine, Campus Box 8108, 660 S Euclid Ave, St Louis, MO, 63110, USA.
| | | | - Heather A Lawson
- Department of Genetics, Washington University School of Medicine, Campus Box 8108, 660 S Euclid Ave, St Louis, MO, 63110, USA.
| |
Collapse
|
22
|
Gourevitch D, Kossenkov AV, Zhang Y, Clark L, Chang C, Showe LC, Heber-Katz E. Inflammation and Its Correlates in Regenerative Wound Healing: An Alternate Perspective. Adv Wound Care (New Rochelle) 2014; 3:592-603. [PMID: 25207202 DOI: 10.1089/wound.2014.0528] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 03/07/2014] [Indexed: 12/21/2022] Open
Abstract
Objective: The wound healing response may be viewed as partially overlapping sets of two physiological processes, regeneration and wound repair with the former overrepresented in some lower species such as newts and the latter more typical of mammals. A robust and quantitative model of regenerative healing has been described in Murphy Roths Large (MRL) mice in which through-and-through ear hole wounds in the ear pinna leads to scarless healing and replacement of all tissue through blastema formation and including cartilage. Since these mice are naturally autoimmune and display many aspects of an enhanced inflammatory response, we chose to examine the inflammatory status during regenerative ear hole closure and observed that inflammation has a clear positive effect on regenerative healing. Approach: The inflammatory gene expression patterns (Illumina microarrays) of early healing ear tissue from regenerative MRL and nonregenerative C57BL/6 (B6) strains are presented along with a survey of innate inflammatory cells found in this tissue type pre and postinjury. The role of inflammation on healing is tested using a COX-2 inhibitor. Innovation and Conclusion: We conclude that (1) enhanced inflammation is consistent with, and probably necessary, for a full regenerative response and (2) the inflammatory gene expression and cell distribution patterns suggest a novel mast cell population with markers found in both immature and mature mast cells that may be a key component of regeneration.
Collapse
Affiliation(s)
| | | | - Yong Zhang
- The Wistar Institute, Philadelphia, Pennsylvania
| | - Lise Clark
- The Wistar Institute, Philadelphia, Pennsylvania
| | - Celia Chang
- The Wistar Institute, Philadelphia, Pennsylvania
| | | | | |
Collapse
|