1
|
Goddard JF, Mehrotra S, Mehrotra M. Osteogenesis imperfecta: exploring an autoimmune and immunotherapy perspective. JBMR Plus 2025; 9:ziaf053. [PMID: 40353205 PMCID: PMC12063996 DOI: 10.1093/jbmrpl/ziaf053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/07/2025] [Accepted: 03/23/2025] [Indexed: 05/14/2025] Open
Abstract
Osteogenesis imperfecta (OI), also called brittle bone disease, is a genetic osteodysplasia characterized by a defect in type 1 collagen. Often diagnosed in infancy or early childhood, young patients are affected by frequent fractures. Osteogenesis imperfecta was first named almost 200 yr ago, yet there are still no FDA-approved treatments for OI, and existing treatments target only the skeletal defects of the disease. In this review, we briefly examine current treatments and ongoing clinical trials. Then, by analyzing OI with an osteoimmunological perspective, we have compiled evidence that OI has an autoimmune component. This autoimmune component of OI remains unconsidered, even though an immunology-based therapy has shown promise in treating OI. Acknowledging an autoimmune component of OI is critical to understanding its mechanisms and allowing for the development of more efficacious treatments and novel immunotherapies. Considering the existing literature and the growing impact of immunotherapeutic therapies in cancer and other autoimmune diseases, we believe it may be time to rethink the immune aspects of this genetic disorder and develop novel immunomodulating strategies to improve the quality of life for OI patients.
Collapse
Affiliation(s)
- Jackson F Goddard
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Meenal Mehrotra
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, United States
| |
Collapse
|
2
|
Sanati M, Pieterman I, Levy N, Akbari T, Tavakoli M, Hassani Najafabadi A, Amin Yavari S. Osteoimmunomodulation by bone implant materials: harnessing physicochemical properties and chemical composition. Biomater Sci 2025; 13:2836-2870. [PMID: 40289736 DOI: 10.1039/d5bm00357a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Chronic inflammation at bone defect sites can impede regenerative processes, but local immune responses can be adjusted to promote healing. Regulating the osteoimmune microenvironment, particularly through macrophage polarization, has become a key focus in bone regeneration research. While bone implants are crucial for addressing significant bone defects, they are often recognized by the immune system as foreign, triggering inflammation that leads to bone resorption and implant issues like fibrous encapsulation and aseptic loosening. Developing osteoimmunomodulatory implants offers a promising approach to transforming destructive inflammation into healing processes, enhancing implant integration and bone regeneration. This review explores strategies based on tuning the physicochemical attributes and chemical composition of materials in engineering osteoimmunomodulatory and pro-regenerative bone implants.
Collapse
Affiliation(s)
- Mehdi Sanati
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands.
| | - Ines Pieterman
- Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Natacha Levy
- Metabolic Diseases Pediatrics Division, University Medical Centre Utrecht, Utrecht, The Netherlands
- Regenerative Medicine Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Tayebeh Akbari
- Department of Microbiology, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | - Mohamadreza Tavakoli
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Saber Amin Yavari
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands.
- Regenerative Medicine Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
3
|
Elazaly H, Dimitriou IM, Maleitzke T, Dahne M, Jaecker V, Maerdian S, Tafelski S, Diekhoff T, Lindner T, Akgün D, Mielke AM, Paksoy A, Amini DA, Planatscher EM, Leopold V, González-Khatib S, Köhli PC, Niemann M, Hildebrandt A, Oehme S, Palmowski Y, Paraskevaidis M, Schönnagel L, Braun SB, Pumberger M, Hardt S, Stricker S, Akyüz L, Grütz G, Schaller S, Lauterbach L, Volcksdorff M, Mödl L, Textor M, Ort M, Reinke S, Stöckle U, Perka C, Duda GN, Schmidt-Bleek K, Geissler S, Winkler T. ILOBONE: A phase I/IIa randomized controlled trial to assess the safety and feasibility of local iloprost therapy for enhancing proximal humerus fracture healing- a pilot study design. J Orthop Surg Res 2025; 20:498. [PMID: 40405317 PMCID: PMC12096472 DOI: 10.1186/s13018-025-05865-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 04/27/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND Proximal humerus fractures (PHFs) are the third most common fractures in elderly patients. Over 70% of PHFs in patients aged over 60 are displaced fractures, often necessitating surgical treatment. However, osteosynthesis is associated with a high rate of complications, highlighting the urgent need for additional therapeutic approaches to enhance bone healing and prevent osteonecrosis. This study evaluates the safety, feasibility and potential efficacy of local prostacyclin (iloprost) to improve bone healing in patients with PHFs. METHODS Thirty eligible patients will be randomized into one of three groups at a 1:1:1 ratio. All patients will receive angular stable locking plate fixation. Two treatment groups will receive an additional single dose of local iloprost through a 24-hour infusion postoperatively (group 1: low dose; group 2: high dose), while the control group will only receive the osteosynthesis. Patients will be monitored for 52 weeks. The primary endpoint is safety, with secondary endpoints including the preservation of the screw tip apex distance as an indicator of fracture healing, head shaft angle, necrosis rate, and patient-related outcome measures. DISCUSSION The Ilobone study aims to provide data on the potential for biological augmentation of osteosynthesis procedures in PHFs, prone to healing challenges and complications. TRIAL REGISTRATION The trial is registered with ClinicalTrial.gov (NCT04543682), registered 02 Sep. 2020, https://clinicaltrials.gov/show/NCT04543682 and the German Clinical Trials Registry (DRKS00027081), registered 10 Nov. 2021 https://drks.de/search/de/trial/DRKS00027081 .
Collapse
Affiliation(s)
- Hisham Elazaly
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Berlin, Germany
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Ioanna Maria Dimitriou
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Berlin, Germany
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Tazio Maleitzke
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Berlin, Germany
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Berlin, Germany
- Department of Orthopedic Surgery, Copenhagen University Hospital- Amager and Hvidovre, Hvidovre, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Michael Dahne
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Vera Jaecker
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sven Maerdian
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sascha Tafelski
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Torsten Diekhoff
- Department of Radiology, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Tobias Lindner
- Emergency Department, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Doruk Akgün
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Anna-Maria Mielke
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Alp Paksoy
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Dominik Adl Amini
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Elisa Marie Planatscher
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Vincent Leopold
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Susana González-Khatib
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Paul Christoph Köhli
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Berlin, Germany
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Junior Clinician Scientist Program, Berlin, Germany
| | - Marcel Niemann
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Alexander Hildebrandt
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Stephan Oehme
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Yannick Palmowski
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Melissa Paraskevaidis
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lukas Schönnagel
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sebastian Benedict Braun
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Matthias Pumberger
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sebastian Hardt
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sigmar Stricker
- Department of Biology, Chemistry and Pharmacy, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Levent Akyüz
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Gerald Grütz
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Stefan Schaller
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Luis Lauterbach
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Berlin, Germany
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Maximilian Volcksdorff
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Berlin, Germany
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Lukas Mödl
- Institute of Biometry and Clinical Epidemiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Martin Textor
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Berlin, Germany
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Melanie Ort
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Berlin, Germany
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Simon Reinke
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Berlin, Germany
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Ulrich Stöckle
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Carsten Perka
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Georg N Duda
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Berlin, Germany
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Berlin, Germany
| | - Katharina Schmidt-Bleek
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Berlin, Germany
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Sven Geissler
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Berlin, Germany
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
- Berlin Center for Advanced Therapies (BECAT), Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Tobias Winkler
- Center for Musculoskeletal Surgery, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Berlin, Germany.
- Berlin Institute of Health (BIH) at Charité- Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany.
- Department of Biology, Chemistry and Pharmacy, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.
- Berlin Center for Advanced Therapies (BECAT), Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
4
|
Häusner S, Kolb A, Übelmesser K, Hölscher-Doht S, Jordan MC, Jauković A, Berberich-Siebelt F, Spasovski DV, Groll J, Blunk T, Herrmann M. It is not waste if it is therapy: cellular, secretory and functional properties of reamer-irrigator-aspirator (RIA)-derived autologous bone grafts. J Orthop Traumatol 2025; 26:21. [PMID: 40140186 PMCID: PMC11947367 DOI: 10.1186/s10195-025-00835-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 03/01/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Large bone defects resulting from trauma, disease, or resection often exceed the intrinsic capacity of bones to heal. The current gold standard addressing these defects is autologous bone grafting (ABG). Procedures such as reamer-irrigator-aspirator (RIA) and conventional bone grafting from the iliac crest are widely recognized as highly effective interventions for critical-size bone defects. The early phase of fracture healing is particularly crucial, as it can determine whether a complete bony union occurs, or if delayed healing or non-unions develop. The initial composition of the bone marrow (BM)-rich ABG transplant, with its unique cellular (e.g., leukocytes, monocytes, and granulocytes) and acellular (e.g., growth factors and extracellular proteins) components, plays a key role in this process. However, despite many successful case reports, the role of ABG cells, growth factors, and their precise contributions to bone healing remain largely elusive. MATERIALS AND METHODS We characterized the native cellularity of both solid and liquid RIA-derived ABG by analyzing primary, minimally manipulated populations of monocytes, macrophages, and T cells, as well as hematopoietic, endothelial, and mesenchymal progenitor cells by flow cytometry. Growth factor and cytokine contents were assessed through antibody arrays. Possible functional and immunomodulatory properties of RIA liquid were evaluated in functional in vitro assays. RESULTS Growth factor and protein arrays revealed a plethora of soluble factors that can be linked to specific immunomodulatory and angiogenic properties, which were evaluated for their potency using functional in vitro assays. We could demonstrate a strong M2-macrophage phenotype inducing the effect of RIA liquid on macrophages. Additionally, we observed an increase in anti-inflammatory T cell subsets generated from peripheral blood mononuclear cells and BM mononuclear cells upon stimulation with RIA liquid . Finally, in vitro endothelial tube formation assays revealed highly significant angiogenic properties of RIA liquid, even at further dilutions. CONCLUSION The cytokine and protein content of RIA liquid exhibits potent immunomodulatory and angiogenic properties. These findings suggest significant therapeutic potential for RIA liquid in modulating immune responses and promoting angiogenesis. Anti-inflammatory and angiogenic properties demonstrated in this study might also help to further define and understand its particular mode of action while also providing explanations to the excellent bone-healing properties of ABG in general. LEVEL OF EVIDENCE Case-series (Level 4).
Collapse
Affiliation(s)
- S Häusner
- Musculoskeletal Cell Biology Group, Institute of Functional Materials and Biofabrication (IFB), University of Würzburg, Röntgenring 11, 97070, Würzburg, Germany.
- Bernhard-Heine-Center for Locomotion Research, Chair of Orthopedics, University of Würzburg, Brettreichstr. 11, 97074, Würzburg, Germany.
| | - A Kolb
- Musculoskeletal Cell Biology Group, Institute of Functional Materials and Biofabrication (IFB), University of Würzburg, Röntgenring 11, 97070, Würzburg, Germany
| | - K Übelmesser
- Musculoskeletal Cell Biology Group, Institute of Functional Materials and Biofabrication (IFB), University of Würzburg, Röntgenring 11, 97070, Würzburg, Germany
| | - S Hölscher-Doht
- Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery (Surgery II), University Hospital Würzburg, Oberdürrbacher Straße 6, 97080, Würzburg, Germany
| | - M C Jordan
- Center for Orthopaedics, Trauma Surgery and Rehabilitation Medicine, University Medicine Greifswald, Fleischmannstraße 8, 17475, Greifswald, Germany
| | - A Jauković
- Group for Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Dr Subotića 4, P.O.B. 102, 11129, Belgrade, Serbia
| | - F Berberich-Siebelt
- Institute of Pathology, University of Würzburg, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - D V Spasovski
- Institute for Orthopedic Surgery (Banjica), University of Belgrade, Milhaila Avramovica 28, Belgrade, Serbia
| | - J Groll
- Department for Functional Materials in Medicine and Dentistry (FMZ), University of Würzburg, Pleicherwall 2, 97070, Würzburg, Germany
| | - T Blunk
- Department of Trauma-, Hand-, Plastic- and Reconstructive Surgery (Surgery II), University Hospital Würzburg, Oberdürrbacher Straße 6, 97080, Würzburg, Germany
| | - M Herrmann
- Musculoskeletal Cell Biology Group, Institute of Functional Materials and Biofabrication (IFB), University of Würzburg, Röntgenring 11, 97070, Würzburg, Germany.
- Bernhard-Heine-Center for Locomotion Research, Chair of Orthopedics, University of Würzburg, Brettreichstr. 11, 97074, Würzburg, Germany.
| |
Collapse
|
5
|
Wang H, Li Y, Li H, Yan X, Jiang Z, Feng L, Hu W, Fan Y, Lin S, Li G. T cell related osteoimmunology in fracture healing: Potential targets for augmenting bone regeneration. J Orthop Translat 2025; 51:82-93. [PMID: 39991456 PMCID: PMC11847249 DOI: 10.1016/j.jot.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/15/2024] [Accepted: 12/01/2024] [Indexed: 02/25/2025] Open
Abstract
UNLABELLED Last decade has witnessed increasing evidence which highlights the roles of immune cells in bone regeneration. Numerous immune cell types, including macrophages, T cells, and neutrophils are involved in fracture healing by orchestrating a series of events that modulate bone formation and remodeling. In this review, the role of T cell immunity in fracture healing has been summarized, and the modulatory effects of T cell immunity in inflammation, bone formation and remodeling have been highlighted. The review also summarizes the specific roles of different T cell subsets, including CD4+ T cells, CD8+ T cells, regulatory T cells, T helper 17 cells, and γδ T cells in modulating fracture healing. The current therapeutics targeting T cell immunity to enhance fracture healing have also been reviewed, aiming to provide insights from a translational standpoint. Overall, this work discusses recent advances and challenges in the interdisciplinary research field of T cell related osteoimmunology and its implications in fracture healing. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE Delayed unions or non-unions of bone fractures remain a challenge in clinical practice. Developing a deep understanding of the roles of immune cells, including T cells, in fracture healing will facilitate the advancement of novel therapeutics of fracture nonunion. This review summarizes the current understanding of different T cell subsets involved in various phases of fracture healing, providing insights for targeting T cells as an alternative strategy to enhance bone regeneration.
Collapse
Affiliation(s)
- Haixing Wang
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yashi Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Haoxin Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xu Yan
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhaowei Jiang
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lu Feng
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, China
| | - Wenhui Hu
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, Guangdong Province, China
| | - Yinuo Fan
- The Third Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Sien Lin
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Gang Li
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
- Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
6
|
Clayton SW, Walk RE, Mpofu L, Easson GWD, Tang SY. Sex-specific divergences in the types and timing of infiltrating immune cells during the intervertebral disc acute injury response and their associations with degeneration. Osteoarthritis Cartilage 2025; 33:247-260. [PMID: 39426787 DOI: 10.1016/j.joca.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 09/17/2024] [Accepted: 10/04/2024] [Indexed: 10/21/2024]
Abstract
OBJECTIVE Inadequate repair of the intervertebral disc (IVD) contributes to low back pain. Infiltrating immune cells into damaged tissues are critical mediators of repair, yet little is known about the identities, roles, and temporal regulation following IVD injury. By analyzing transcripts of immune cell markers, histopathologic analysis, immunofluorescence, and flow cytometry, we aimed to define the temporal cascade of infiltrating immune cells and their associations with IVD degeneration. METHODS Caudal IVDs from 12-week-old C57BL6/J mice were injured and monitored for 42 days post-injury. Transcriptional markers identifying myeloid, B, and T immune cells, and angiogenic factors were measured from the IVDs every 2-3 days. Histopathologic degeneration of the IVD was measured throughout. Flow cytometry and immunofluorescence were used to identify and localize cells including B, T, natural killer T (NKT) cells, monocytes, neutrophils, macrophages, eosinophils, and dendritic cells. RESULTS The injured IVD revealed distinct phases of inflammation and proliferation. Robust temporal oscillation in the myeloid and T cell transcripts was observed in females. Cd3+ T cells were more abundant in females than in males. The Cd3+Cd4-Cd8- T cells that dominate the female cascade contain rare γδ T cells. Injury-mediated degeneration was prevalent in both sexes but more severe in males. CONCLUSIONS This study defines the coordinated infiltration of immune cells in the IVD following injury. We report the discovery of γδ T cells in the female IVD, and this was associated with less severe degeneration. γδ T cells have potent anti-inflammatory roles and may suppress degeneration following IVD injury.
Collapse
Affiliation(s)
| | - Remy E Walk
- Washington University in St. Louis, St. Louis, MO, USA
| | - Laura Mpofu
- Washington University in St. Louis, St. Louis, MO, USA
| | | | - Simon Y Tang
- Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
7
|
Ahn J, Kim B, Bello AB, Moon JJ, Arai Y, Lee SH. Regenerative Functions of Regulatory T Cells and Current Strategies Utilizing Mesenchymal Stem Cells in Immunomodulatory Tissue Regeneration. Tissue Eng Regen Med 2025; 22:167-180. [PMID: 39804546 PMCID: PMC11794763 DOI: 10.1007/s13770-024-00690-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: 11/21/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Regulatory T cells (Tregs) are essential for maintaining immune homeostasis and facilitating tissue regeneration by fostering an environment conducive to tissue repair. However, in damaged tissues, excessive inflammatory responses can overwhelm the immunomodulatory capacity of Tregs, compromising their functionality and potentially hindering effective regeneration. Mesenchymal stem cells (MSCs) play a key role in enhancing Treg function. MSCs enhance Treg activity through indirect interactions, such as cytokine secretion, and direct interactions via membrane proteins. METHODS This review examines the regenerative functions of Tregs across various tissues, including bone, cartilage, muscle, and skin, and explores strategies to enhance Treg functionality using MSCs. Advanced techniques, such as the overexpression of relevant genes in MSCs, are highlighted for their potential to further enhance Treg function. Additionally, emerging technologies utilizing extracellular vesicles (EVs) and cell membrane-derived vesicles derived from MSCs offer promising alternatives to circumvent the potential side effects associated with live cell therapies. This review proposes approaches to enhance Treg function and promote tissue regeneration and also outlines future research directions. RESULTS AND CONCLUSION This review elucidates recent technological advancements aimed at enhancing Treg function using MSCs and examines their potential to improve tissue regeneration efficiency.
Collapse
Grants
- 2022R1A2C3004850 Ministry of Science and ICT, South Korea
- RS-2024-00405381 Ministry of Science and ICT, South Korea
- RS-2023-00257290 Ministry of Science and ICT, South Korea
- RS-2023-00246418 Ministry of Education
- RS-2023-00275407 Ministry of Education
- 21C0703L1 Ministry of Science and ICT, Ministry of Health & Welfare
- HX23C1734 Ministry of Science and ICT, Ministry of Trade, Industry and Energy, Ministry of Health & Welfare, The Ministry of Food and Drug Safety
- Ministry of Science and ICT, Ministry of Health & Welfare
- Ministry of Science and ICT, Ministry of Trade, Industry and Energy, Ministry of Health & Welfare, The Ministry of Food and Drug Safety
Collapse
Affiliation(s)
- Jinsung Ahn
- Department of Biomedical Engineering, Dongguk University, Seoul, South Korea
| | - Bowon Kim
- Department of Biomedical Engineering, Dongguk University, Seoul, South Korea
| | - Alvin Bacero Bello
- Department of Biomedical Engineering, Dongguk University, Seoul, South Korea
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Yoshie Arai
- Department of Biomedical Engineering, Dongguk University, Seoul, South Korea.
| | - Soo-Hong Lee
- Department of Biomedical Engineering, Dongguk University, Seoul, South Korea.
| |
Collapse
|
8
|
Burgan J, Rahmati M, Lee M, Saiz AM. Innate immune response to bone fracture healing. Bone 2025; 190:117327. [PMID: 39522707 DOI: 10.1016/j.bone.2024.117327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
The field of osteoimmunology has primarily focused on fracture healing in isolated musculoskeletal injuries. The innate immune system is the initial response to fracture, with inflammatory macrophages, cytokines, and neutrophils arriving first at the fracture hematoma, followed by an anti-inflammatory phase to begin the process of new bone formation. This review aims to first discuss the current literature and knowledge gaps on the immune responses governing single fracture healing by encompassing the individual role of macrophages, neutrophils, cytokines, mesenchymal stem cells, bone cells, and other immune cells. This paper discusses the interactive effects of these cellular responses underscoring the field of osteoimmunology. The critical role of the metabolic environment in guiding the immune system properties will be highlighted along with some effective therapeutics for fracture healing in the context of osteoimmunology. However, compared to isolated fractures, which frequently heal well, long bone fractures in over 30 % of polytrauma patients exhibit impaired healing. Clinical evidence suggests there may be distinct physiologic and inflammatory pathways altered in polytrauma resulting in nonunion. Nonunion is associated with worse patient outcomes and increased societal healthcare costs. The dysregulated immunomodulatory/inflammatory response seen in polytrauma may lead to this increased nonunion rate. This paper will investigate the differences in immune response between isolated and polytrauma fractures. Finally, future directions for fracture studies are explored with consideration of the emerging roles of newly discovered immune cell functions in fracture healing, the existing challenges and conflicting results in the field, the translational potential of these studies in clinic, and the more complex nature of polytrauma fractures that can alter cell functions in different tissues.
Collapse
Affiliation(s)
- Jane Burgan
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA; Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Maryam Rahmati
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA; Department of Biomaterials, Institute for Clinical Dentistry, University of Oslo, PO Box 1109, Blindern, NO-0317 Oslo, Norway
| | - Mark Lee
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA
| | - Augustine Mark Saiz
- Department of Orthopaedic Surgery, UC Davis Health, 4860 Y Street, Suite 3800, Sacramento, CA 95817, USA.
| |
Collapse
|
9
|
Graef F, Wei Y, Garbe A, Seemann R, Zenzes M, Tsitsilonis S, Duda GN, Zaslansky P. Increased cancellous bone mass accompanies decreased cortical bone mineral density and higher axial deformation in femurs of leptin-deficient obese mice. J Mech Behav Biomed Mater 2024; 160:106745. [PMID: 39317095 DOI: 10.1016/j.jmbbm.2024.106745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 09/02/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024]
Abstract
INTRODUCTION Leptin is a pleiotropic hormone that regulates food intake and energy homeostasis with enigmatic effects on bone development. It is unclear if leptin promotes or inhibits bone growth. The aim of this study was to characterize the micro-architecture and mechanical competence of femur bones of leptin-deficient mice. MATERIALS AND METHODS Right femur bones of 15-week old C57BL/6 (n = 9) and leptin-deficient (ob/ob, n = 9) mice were analyzed. Whole bones were scanned using micro-CT and morphometric parameters of the cortex and trabeculae were assessed. Elastic moduli were determined from microindentations in midshaft cross-sections. Mineral densities were determined using quantitative backscatter scanning electron microscopy. 3D models of the distal femur metaphysis, cleared from trabecular bone, were meshed and used for finite element simulations of axial loading to identify straining differences between ob/ob and C57BL/6 controls. RESULTS Compared with C57BL/6 controls, ob/ob mice had significantly shorter bones. ob/ob mice showed significantly increased cancellous bone volume and trabecular thickness. qBEI quantified a ∼7% lower mineral density in ob/ob mice in the distal femur metaphysis. Indentation demonstrated a significantly reduced Young's modulus of 12.14 [9.67, 16.56 IQR] GPa for ob/ob mice compared to 23.12 [20.70, 26.57 IQR] GPa in C57BL/6 mice. FEA revealed greater deformation of cortical bone in ob/ob as compared to C57BL/6 mice. CONCLUSION Leptin deficient ob/ob mice have a softer cortical bone in the distal femur metaphysis but an excessive amount of cancellous bone, possibly as a response to increased deformation of the bones during axial loading. Both FEA and direct X-ray and electron microscopy imaging suggest that the morphology and micro-architecture of ob/ob mice have inferior biomechanical properties suggestive of a reduced mechanical competence.
Collapse
Affiliation(s)
- F Graef
- Charité - Universitätsmedizin Berlin, Center for Musculoskeletal Surgery, Germany; Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany.
| | - Y Wei
- Charité - Universitätsmedizin Berlin, Department of Operative and Preventive Dentistry, Germany; Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany.
| | - A Garbe
- Charité - Universitätsmedizin Berlin, Center for Musculoskeletal Surgery, Germany
| | - R Seemann
- Charité - Universitätsmedizin Berlin, Center for Musculoskeletal Surgery, Germany
| | - M Zenzes
- Charité - Universitätsmedizin Berlin, Department of Operative and Preventive Dentistry, Germany
| | - S Tsitsilonis
- Charité - Universitätsmedizin Berlin, Center for Musculoskeletal Surgery, Germany; Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany
| | - G N Duda
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany
| | - P Zaslansky
- Charité - Universitätsmedizin Berlin, Department of Operative and Preventive Dentistry, Germany.
| |
Collapse
|
10
|
Voss JO, Pivetta F, Elkilany A, Schmidt-Bleek K, Duda GN, Odaka K, Dimitriou IM, Ort MJ, Streitz M, Heiland M, Koerdt S, Reinke S, Geissler S. Prognostic implications of a CD8 + T EMRA to CD4 +T reg imbalance in mandibular fracture healing: a prospective analysis of immune profiles. Front Immunol 2024; 15:1476009. [PMID: 39507538 PMCID: PMC11537918 DOI: 10.3389/fimmu.2024.1476009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 10/08/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction Open reduction and fixation are the standard of care for treating mandibular fractures and usually lead to successful healing. However, complications such as delayed healing, non-union, and infection can compromise patient outcomes and increase healthcare costs. The initial inflammatory response, particularly the response involving specific CD8+ T cell subpopulations, is thought to play a critical role in healing long bone fractures. In this study, we investigated the role of these immune cell profiles in patients with impaired healing of mandibular fractures. Materials and methods In this prospective study, we included patients with mandibular fractures surgically treated at Charité - Universitätsmedizin Berlin, Germany, between September 2020 and December 2022. We used follow-up imaging and clinical assessment to evaluate bone healing. In addition, we analyzed immune cell profiles using flow cytometry and quantified cytokine levels using electrochemiluminescence-based multiplex immunoassays in preoperative blood samples. Results Out of the 55 patients enrolled, 38 met the inclusion criteria (30 men and 8 women; mean age 32.18 years). Radiographic evaluation revealed 31 cases of normal healing and 7 cases of incomplete consolidation, including 1 case of non-union. Patients with impaired healing exhibited increased levels of terminally differentiated effector memory CD8+ T cells (TEMRA) and a higher TEMRA to regulatory T cell (Treg) ratio, compared with those with normal healing. Conclusions Our analysis of mandibular fracture cases confirms our initial hypothesis derived from long bone fracture healing: monitoring the TEMRA to Treg ratio in preoperative blood can be an early indicator of patients at risk of impaired bone healing. Radiologic follow-up enabled us to detect healing complications that might not be detected by clinical assessment only. This study highlights the potential of individual immune profiles to predict successful healing and may form the basis for future strategies to manage healing complications.
Collapse
Affiliation(s)
- Jan Oliver Voss
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Oral and Maxillofacial Surgery, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Academy, Clinician Scientist Program, Berlin, Germany
| | - Fabio Pivetta
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Department of Radiology, Berlin, Germany
| | - Aboelyazid Elkilany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Department of Radiology, Berlin, Germany
| | - Katharina Schmidt-Bleek
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Berlin, Germany
- Berlin Institute of Health at Charité Universitätsmedizin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Georg N. Duda
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Berlin, Germany
- Berlin Institute of Health at Charité Universitätsmedizin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Kento Odaka
- Department of Oral and Maxillofacial Radiology, Tokyo Dental College, Chiyoda-Ku, Tokyo, Japan
| | - Ioanna Maria Dimitriou
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Berlin, Germany
- Berlin Institute of Health at Charité Universitätsmedizin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
| | - Melanie Jasmin Ort
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Berlin, Germany
- Berlin Institute of Health at Charité Universitätsmedizin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
| | - Mathias Streitz
- Friedrich-Loeffler-Institute, Federal Research Institute for Animal Health, Greifswald – Insel Riems, Germany
| | - Max Heiland
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Oral and Maxillofacial Surgery, Berlin, Germany
| | - Steffen Koerdt
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Oral and Maxillofacial Surgery, Berlin, Germany
| | - Simon Reinke
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Berlin, Germany
- Berlin Institute of Health at Charité Universitätsmedizin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Sven Geissler
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Berlin, Germany
- Berlin Institute of Health at Charité Universitätsmedizin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| |
Collapse
|
11
|
Sun T, Li C, Luan J, Zhao F, Zhang Y, Liu J, Shao L. Black phosphorus for bone regeneration: Mechanisms involved and influencing factors. Mater Today Bio 2024; 28:101211. [PMID: 39280114 PMCID: PMC11402231 DOI: 10.1016/j.mtbio.2024.101211] [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: 05/18/2024] [Revised: 08/19/2024] [Accepted: 08/23/2024] [Indexed: 09/18/2024] Open
Abstract
BP has shown good potential for promoting bone regeneration. However, the understanding of the mechanisms of BP-enhanced bone regeneration is still limited. This review first summarizes the recent advances in applications of BP in bone regeneration. We further highlight the possibility that BP enhances bone regeneration by regulating the behavior of mesenchymal stem cells (MSCs), osteoblasts, vascular endothelial cells (VECs), and macrophages, mainly through the regulation of cytoskeletal remodeling, energy metabolism, oxidation resistance and surface adsorption properties, etc. In addition, moderating the physicochemical properties of BP (i.e., shape, size, and surface charge) can alter the effects of BP on bone regeneration. This review reveals the underlying mechanisms of BP-enhanced bone regeneration and provides strategies for further material design of BP-based materials for bone regeneration.
Collapse
Affiliation(s)
- Ting Sun
- Foshan Stomatology Hospital & School of Medicine, Foshan University, Foshan, 528000, China
- School of Dentistry, Jinan University, Guangzhou, 510630, China
| | - Chufeng Li
- School of Dentistry, Jinan University, Guangzhou, 510630, China
| | - Jiayi Luan
- Foshan Stomatology Hospital & School of Medicine, Foshan University, Foshan, 528000, China
| | - Fujian Zhao
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yanli Zhang
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Jia Liu
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Longquan Shao
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| |
Collapse
|
12
|
Siverino C, Metsemakers WJ, Sutter R, Della Bella E, Morgenstern M, Barcik J, Ernst M, D'Este M, Joeris A, Chittò M, Schwarzenberg P, Stoddart M, Vanvelk N, Richards G, Wehrle E, Weisemann F, Zeiter S, Zalavras C, Varga P, Moriarty TF. Clinical management and innovation in fracture non-union. Expert Opin Biol Ther 2024; 24:973-991. [PMID: 39126182 DOI: 10.1080/14712598.2024.2391491] [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: 05/21/2024] [Revised: 07/18/2024] [Accepted: 08/08/2024] [Indexed: 08/12/2024]
Abstract
INTRODUCTION With the introduction and continuous improvement in operative fracture fixation, even the most severe bone fractures can be treated with a high rate of successful healing. However, healing complications can occur and when healing fails over prolonged time, the outcome is termed a fracture non-union. Non-union is generally believed to develop due to inadequate fixation, underlying host-related factors, or infection. Despite the advancements in fracture fixation and infection management, there is still a clear need for earlier diagnosis, improved prediction of healing outcomes and innovation in the treatment of non-union. AREAS COVERED This review provides a detailed description of non-union from a clinical perspective, including the state of the art in diagnosis, treatment, and currently available biomaterials and orthobiologics.Subsequently, recent translational development from the biological, mechanical, and infection research fields are presented, including the latest in smart implants, osteoinductive materials, and in silico modeling. EXPERT OPINION The first challenge for future innovations is to refine and to identify new clinical factors for the proper definition, diagnosis, and treatment of non-union. However, integration of in vitro, in vivo, and in silico research will enable a comprehensive understanding of non-union causes and correlations, leading to the development of more effective treatments.
Collapse
Affiliation(s)
- C Siverino
- AO Research Institute Davos, Davos Platz, Switzerland
| | - W-J Metsemakers
- Department of Trauma Surgery, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven - University of Leuven, Leuven, Belgium
| | - R Sutter
- Radiology Department, Balgrist University Hospital, University of Zürich, Zürich, Switzerland
| | - E Della Bella
- AO Research Institute Davos, Davos Platz, Switzerland
| | - M Morgenstern
- Center for Musculoskeletal Infections, Department of Orthopaedic and Trauma Surgery, University Hospital Basel, Basel, Switzerland
| | - J Barcik
- AO Research Institute Davos, Davos Platz, Switzerland
| | - M Ernst
- AO Research Institute Davos, Davos Platz, Switzerland
| | - M D'Este
- AO Research Institute Davos, Davos Platz, Switzerland
| | - A Joeris
- AO Innovation Translation Center, Davos Platz, Switzerland
| | - M Chittò
- AO Research Institute Davos, Davos Platz, Switzerland
| | | | - M Stoddart
- AO Research Institute Davos, Davos Platz, Switzerland
| | - N Vanvelk
- Trauma Research Unit, Department of Surgery, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - G Richards
- AO Research Institute Davos, Davos Platz, Switzerland
| | - E Wehrle
- AO Research Institute Davos, Davos Platz, Switzerland
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - F Weisemann
- Department of Trauma Surgery, BG Unfallklinik Murnau, Murnau am Staffelsee, Germany
| | - S Zeiter
- AO Research Institute Davos, Davos Platz, Switzerland
| | - C Zalavras
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - P Varga
- AO Research Institute Davos, Davos Platz, Switzerland
| | - T F Moriarty
- AO Research Institute Davos, Davos Platz, Switzerland
- Center for Musculoskeletal Infections, Department of Orthopaedic and Trauma Surgery, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
13
|
Shi W, Feng Y, Tang J, Xu Y, Wang W, Zhang L, Jiang X, Ding Z, Xi K, Chen L, Gu Y. A Genetically Engineered "Reinforced Concrete" Scaffold Regulates the N2 Neutrophil Innate Immune Cascade to Repair Bone Defects. Adv Healthc Mater 2024; 13:e2304585. [PMID: 38411324 DOI: 10.1002/adhm.202304585] [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: 12/22/2023] [Revised: 02/17/2024] [Indexed: 02/28/2024]
Abstract
The innate immune response is crucial to inflammation, but how neutrophils and macrophages act in bone repair and tissue engineering treatment strategies await clarification. In this study, it is found that N2 neutrophils release stronger "eat me" signals to induce macrophage phagocytosis and polarize into the M2 anti-inflammatory phenotype. Guided by this biological mechanism, a mesoporous bioactive glass scaffold (MBG) is filled with hyaluronic acid methacryloyl (HAMA) hydrogel loaded with Transforming growth factor-β1 (TGFβ1) adenovirus (Ad@H), constructing a genetically engineered composite scaffold (Ad@H/M). The scaffold not only has good hydrophilicity and biocompatibility, but also provides mechanical stress support for bone repair. Adenovirus infection quickly induces N2 neutrophils, upregulating NF-κB and MAPK signaling pathways through Toll-like receptor 4 (TLR4) to promote the inflammatory response and macrophage phagocytosis. Macrophages perform phagocytosis and polarize towards the M2 phenotype, mediating the inflammatory response by inhibiting the PI3K-AKT-NF-κB pathway, maintaining homeostasis of the osteogenic microenvironment. The role of the Ad@H/M scaffold in regulating early inflammation and promoting long-term bone regeneration is further validated in vivo. In brief, this study focuses on the cascade of reactions between neutrophils and macrophage subtypes, and reports a composite scaffold that coordinates the innate immune response to promote bone repair.
Collapse
Affiliation(s)
- Wenxiao Shi
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Yu Feng
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Jincheng Tang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Yichang Xu
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Wei Wang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Lichen Zhang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Xinzhao Jiang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Zhouye Ding
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Kun Xi
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Liang Chen
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| | - Yong Gu
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, P. R. China
| |
Collapse
|
14
|
Han J, Rindone AN, Elisseeff JH. Immunoengineering Biomaterials for Musculoskeletal Tissue Repair across Lifespan. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311646. [PMID: 38416061 PMCID: PMC11239302 DOI: 10.1002/adma.202311646] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/23/2024] [Indexed: 02/29/2024]
Abstract
Musculoskeletal diseases and injuries are among the leading causes of pain and morbidity worldwide. Broad efforts have focused on developing pro-regenerative biomaterials to treat musculoskeletal conditions; however, these approaches have yet to make a significant clinical impact. Recent studies have demonstrated that the immune system is central in orchestrating tissue repair and that targeting pro-regenerative immune responses can improve biomaterial therapeutic outcomes. However, aging is a critical factor negatively affecting musculoskeletal tissue repair and immune function. Hence, understanding how age affects the response to biomaterials is essential for improving musculoskeletal biomaterial therapies. This review focuses on the intersection of the immune system and aging in response to biomaterials for musculoskeletal tissue repair. The article introduces the general impacts of aging on tissue physiology, the immune system, and the response to biomaterials. Then, it explains how the adaptive immune system guides the response to injury and biomaterial implants in cartilage, muscle, and bone and discusses how aging impacts these processes in each tissue type. The review concludes by highlighting future directions for the development and translation of personalized immunomodulatory biomaterials for musculoskeletal tissue repair.
Collapse
Affiliation(s)
- Jin Han
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University; Baltimore, MD 21231, USA
| | - Alexandra N. Rindone
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University; Baltimore, MD 21231, USA
| | - Jennifer H. Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University; Baltimore, MD 21231, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine; Baltimore, MD 21231, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University; Baltimore, MD 21231, USA
| |
Collapse
|
15
|
Molitoris KH, Balu AR, Huang M, Baht GS. The impact of age and sex on the inflammatory response during bone fracture healing. JBMR Plus 2024; 8:ziae023. [PMID: 38560342 PMCID: PMC10978063 DOI: 10.1093/jbmrpl/ziae023] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 12/29/2023] [Accepted: 02/12/2024] [Indexed: 04/04/2024] Open
Abstract
Inflammation is thought to be dysregulated with age leading to impaired bone fracture healing. However, broad analyses of inflammatory processes during homeostatic bone aging and during repair are lacking. Here, we assessed changes in inflammatory cell and cytokine profiles in circulation and in bone tissue to identify age- and sex-dependent differences during homeostasis and repair. During homeostatic aging, male mice demonstrated accumulation of CD4+ helper T cells and CD8+ cytotoxic T cells within bone while both pro-inflammatory "M1" and anti-inflammatory "M2" macrophage numbers decreased. Female mice saw no age-associated changes in immune-cell population in homeostatic bone. Concentrations of IL-1β, IL-9, IFNγ, and CCL3/MIP-1α increased with age in both male and female mice, whereas concentrations of IL-2, TNFα, TNFR1, IL-4, and IL-10 increased only in female mice - thus we termed these "age-accumulated" cytokines. There were no notable changes in immune cell populations nor cytokines within circulation during aging. Sex-dependent analysis demonstrated slight changes in immune cell and cytokine levels within bone and circulation, which were lost upon fracture injury. Fracture in young male mice caused a sharp decrease in number of M1 macrophages; however, this was not seen in aged male mice nor in female mice of any age. Injury itself induced a decrease in the number of CD8+ T cells within the local tissue of aged male and of female mice but not of young mice. Cytokine analysis of fractured mice revealed that age-accumulated cytokines quickly dissipated after fracture injury, and did not re-accumulate in newly regenerated tissue. Conversely, CXCL1/KC-GRO, CXCL2/MIP-2, IL-6, and CCL2/MCP-1 acted as "fracture response" cytokines: increasing sharply after fracture, eventually returning to baseline. Collectively, we classify measured cytokines into three groups: (1) age-accumulated cytokines, (2) female-specific age-accumulated cytokines, and (3) fracture response cytokines. These inflammatory molecules represent potential points of intervention to improve fracture healing outcome.
Collapse
Affiliation(s)
- Kristin Happ Molitoris
- Department of Orthopaedic Surgery, Duke Molecular Physiology Institute, Department of Pathology, Duke University, Durham, NC 27701, United States
| | - Abhinav Reddy Balu
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Mingjian Huang
- Department of Orthopaedic Surgery, Duke Molecular Physiology Institute, Department of Pathology, Duke University, Durham, NC 27701, United States
| | - Gurpreet Singh Baht
- Department of Orthopaedic Surgery, Duke Molecular Physiology Institute, Department of Pathology, Duke University, Durham, NC 27701, United States
| |
Collapse
|
16
|
Avery D, Morandini L, Sheakley L, Grabiec M, Olivares-Navarrete R. CD4 + and CD8 + T cells reduce inflammation and promote bone healing in response to titanium implants. Acta Biomater 2024; 179:385-397. [PMID: 38554889 PMCID: PMC11045310 DOI: 10.1016/j.actbio.2024.03.022] [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: 11/27/2023] [Revised: 03/11/2024] [Accepted: 03/24/2024] [Indexed: 04/02/2024]
Abstract
T cells are adaptive immune cells essential in pathogenic response, cancer, and autoimmune disorders. During the integration of biomaterials with host tissue, T cells modify the local inflammatory environment by releasing cytokines that promote inflammatory resolution following implantation. T cells are vital for the modulation of innate immune cells, recruitment and proliferation of mesenchymal stem cells (MSCs), and formation of functional tissue around the biomaterial implant. We have demonstrated that deficiency of αβ T cells promotes macrophage polarization towards a pro-inflammatory phenotype and attenuates MSC recruitment and proliferation in vitro and in vivo. The goal of this study was to understand how CD4+ and CD8+ T cells, subsets of the αβ T cell family, impact the inflammatory response to titanium (Ti) biomaterials. Deficiency of either CD4+ or CD8+ T cells increased the proportion of pro-inflammatory macrophages, lowered anti-inflammatory macrophages, and diminished MSC recruitment in vitro and in vivo. In addition, new bone formation at the implantation site was significantly reduced in T cell-deficient mice compared to T cell-competent mice. Deficiency of CD4+ T cells exacerbated these effects compared to CD8+ T cell deficiency. Our results show the importance of CD4+ and CD8+ T cells in modulating the inflammatory response and promoting new bone formation in response to modified Ti implants. STATEMENT OF SIGNIFICANCE: CD4+ and CD8+ T cells are essential in modulating the peri-implant microenvironment during the inflammatory response to biomaterial implantation. This study shows that deficiency of either CD4+ or CD8+ T cell subsets altered macrophage polarization and reduced MSC recruitment and proliferation at the implantation site.
Collapse
Affiliation(s)
- Derek Avery
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 70 S. Madison Street, Room 3328, Richmond, VA 23220, United States
| | - Lais Morandini
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 70 S. Madison Street, Room 3328, Richmond, VA 23220, United States
| | - Luke Sheakley
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 70 S. Madison Street, Room 3328, Richmond, VA 23220, United States
| | - Melissa Grabiec
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 70 S. Madison Street, Room 3328, Richmond, VA 23220, United States
| | - Rene Olivares-Navarrete
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, 70 S. Madison Street, Room 3328, Richmond, VA 23220, United States.
| |
Collapse
|
17
|
Palomares Cabeza V, Fahy N, Kiernan CH, Lolli A, Witte-Bouma J, Fahmy Garcia S, Merino A, Kops N, Ridwan Y, Wolvius EB, Brama PAJ, Hoogduijn MJ, Farrell E. Bone formation by human paediatric marrow stromal cells in a functional allogeneic immune system. Biomaterials 2024; 306:122471. [PMID: 38377846 DOI: 10.1016/j.biomaterials.2024.122471] [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: 02/22/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 02/22/2024]
Abstract
Allogeneic stem-cell based regenerative medicine is a promising approach for bone defect repair. The use of chondrogenically differentiated human marrow stromal cells (MSCs) has been shown to lead to bone formation by endochondral ossification in immunodeficient pre-clinical models. However, an insight into the interactions between the allogeneic immune system and the human MSC-derived bone grafts has not been fully achieved yet. The choice of a potent source of MSCs isolated from pediatric donors with consistent differentiation and high proliferation abilities, as well as low immunogenicity, could increase the chance of success for bone allografts. In this study, we employed an immunodeficient animal model humanised with allogeneic immune cells to study the immune responses towards chondrogenically differentiated human pediatric MSCs (ch-pMSCs). We show that ch-differentiated pMSCs remained non-immunogenic to allogeneic CD4 and CD8 T cells in an in vitro co-culture model. After subcutaneous implantation in mice, ch-pMSC-derived grafts were able to initiate bone mineralisation in the presence of an allogeneic immune system for 3 weeks without the onset of immune responses. Re-exposing the splenocytes of the humanised animals to pMSCs did not trigger further T cell proliferation, suggesting an absence of secondary immune responses. Moreover, ch-pMSCs generated mature bone after 8 weeks of implantation that persisted for up to 6 more weeks in the presence of an allogeneic immune system. These data collectively show that human allogeneic chondrogenically differentiated pediatric MSCs might be a safe and potent option for bone defect repair in the tissue engineering and regenerative medicine setting.
Collapse
Affiliation(s)
- Virginia Palomares Cabeza
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Niamh Fahy
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Orthopedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Applied Science, Technological University of the Shannon: Midlands Midwest, Limerick, Ireland
| | - Caoimhe H Kiernan
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Andrea Lolli
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Janneke Witte-Bouma
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Shorouk Fahmy Garcia
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Orthopedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Ana Merino
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Nicole Kops
- Department of Orthopedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Yanto Ridwan
- AMIE Core Facility, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Eppo B Wolvius
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Pieter A J Brama
- School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Martin J Hoogduijn
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| |
Collapse
|
18
|
Clayton SW, Walk RE, Mpofu L, Easson GW, Tang SY. Analysis of Infiltrating Immune Cells Following Intervertebral Disc Injury Reveals Recruitment of Gamma-Delta ( γδ) T cells in Female Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.01.582950. [PMID: 38464124 PMCID: PMC10925253 DOI: 10.1101/2024.03.01.582950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Inadequate repair of injured intervertebral discs (IVD) leads to degeneration and contributes to low back pain. Infiltrating immune cells into damaged musculoskeletal tissues are critical mediators of repair, yet little is known about their identities, roles, and temporal regulation following IVD injury. By analyzing longitudinal changes in gene expression, tissue morphology, and the dynamics of infiltrating immune cells following injury, we characterize sex-specific differences in immune cell populations and identify the involvement of previously unreported immune cell types, γδ and NKT cells. Cd3+Cd4-Cd8- T cells are the largest infiltrating lymphocyte population with injury, and we identified the presence of γδ T cells in this population in female mice specifically, and NKT cells in males. Injury-mediated IVD degeneration was prevalent in both sexes, but more severe in males. Sex-specific degeneration may be associated with the differential immune response since γδ T cells have potent anti-inflammatory roles and may mediate IVD repair.
Collapse
Affiliation(s)
| | - Remy E. Walk
- Washington University in St. Louis, St. Louis, MO
| | - Laura Mpofu
- Washington University in St. Louis, St. Louis, MO
| | | | | |
Collapse
|
19
|
Li J, Xia T, Zhao Q, Wang C, Fu L, Zhao Z, Tang Z, Yin C, Wang M, Xia H. Biphasic calcium phosphate recruits Tregs to promote bone regeneration. Acta Biomater 2024; 176:432-444. [PMID: 38185232 DOI: 10.1016/j.actbio.2024.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 12/27/2023] [Accepted: 01/01/2024] [Indexed: 01/09/2024]
Abstract
The use of bone substitute materials is crucial for the healing of large bone defects. Immune response induced by bone substitute materials is essential in bone regeneration. Prior research has mainly concentrated on innate immune cells, such as macrophages. Existing research suggests that T lymphocytes, as adaptive immune cells, play an indispensable role in bone regeneration. However, the mechanisms governing T cell recruitment and specific subsets that are essential for bone regeneration remain unclear. This study demonstrates that CD4+ T cells are indispensable for ectopic osteogenesis by biphasic calcium phosphate (BCP). Subsequently, the recruitment of CD4+ T cells is closely associated with the activation of calcium channels in macrophages by BCP to release chemokines Ccl3 and Ccl17. Finally, these recruited CD4+ T cells are predominantly Tregs, which play a significant role in ectopic osteogenesis by BCP. These findings not only shed light on the immune-regenerative process after bone substitute material implantation but also establish a theoretical basis for developing bone substitute materials for promoting bone tissue regeneration. STATEMENT OF SIGNIFICANCE: Bone substitute material implantation is essential in the healing of large bone defects. Existing research suggests that T lymphocytes are instrumental in bone regeneration. However, the specific mechanisms governing T cell recruitment and specific subsets that are essential for bone regeneration remain unclear. In this study, we demonstrate that activation of calcium channels in macrophages by biphasic calcium phosphate (BCP) causes them to release the chemokines Ccl3 and Ccl17 to recruit CD4+ T cells, predominantly Tregs, which play a crucial role in ectopic osteogenesis by BCP. Our findings provide a theoretical foundation for developing bone substitute material for bone tissue regeneration.
Collapse
Affiliation(s)
- Jiaojiao Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Ting Xia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Qin Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Can Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Liangliang Fu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zifan Zhao
- Center of Digital Dentistry, Faculty of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & NHC Key Laboratory of Digital Stomatology & Beijing Key Laboratory of Digital Stomatology & Key Laboratory of Digital Stomatology, Chinese Academy of Medical Sciences & NMPA Key Laboratory for Dental Materials, Beijing,100081, China
| | - Ziqiao Tang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Chenghu Yin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Min Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Haibin Xia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| |
Collapse
|
20
|
Qiu M, Tulufu N, Tang G, Ye W, Qi J, Deng L, Li C. Black Phosphorus Accelerates Bone Regeneration Based on Immunoregulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304824. [PMID: 37953457 PMCID: PMC10767454 DOI: 10.1002/advs.202304824] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/25/2023] [Indexed: 11/14/2023]
Abstract
A fundamental understanding of inflammation and tissue healing suggests that the precise regulation of the inflammatory phase, both in terms of location and timing, is crucial for bone regeneration. However, achieving the activation of early inflammation without causing chronic inflammation while facilitating quick inflammation regression to promote bone regeneration continues to pose challenges. This study reveals that black phosphorus (BP) accelerates bone regeneration by building an osteogenic immunological microenvironment. BP amplifies the acute pro-inflammatory response and promotes the secretion of anti-inflammatory factors to accelerate inflammation regression and tissue regeneration. Mechanistically, BP creates an osteoimmune-friendly microenvironment by stimulating macrophages to express interleukin 33 (IL-33), amplifying the inflammatory response at an early stage, and promoting the regression of inflammation. In addition, BP-mediated IL-33 expression directly promotes osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), which further facilitates bone repair. To the knowledge, this is the first study to reveal the immunomodulatory potential of BP in bone regeneration through the regulation of both early-stage inflammatory responses and later-stage inflammation resolution, along with the associated molecular mechanisms. This discovery serves as a foundation for the clinical use of BP and is an efficient approach for managing the immune microenvironment during bone regeneration.
Collapse
Affiliation(s)
- Minglong Qiu
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Nijiati Tulufu
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Guoqing Tang
- Kunshan Hospital of Traditional Chinese MedicineAffiliated Hospital of Yangzhou University388 Zuchongzhi RoadKunshan CityJiangsu Province215300P. R. China
| | - Wenkai Ye
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Jin Qi
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Lianfu Deng
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Changwei Li
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| |
Collapse
|
21
|
Schlundt C, Saß RA, Bucher CH, Bartosch S, Hauser AE, Volk HD, Duda GN, Schmidt-Bleek K. Complex Spatio-Temporal Interplay of Distinct Immune and Bone Cell Subsets during Bone Fracture Healing. Cells 2023; 13:40. [PMID: 38201244 PMCID: PMC10777943 DOI: 10.3390/cells13010040] [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: 11/10/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND The healing of a bone injury is a highly complex process involving a multitude of different tissue and cell types, including immune cells, which play a major role in the initiation and progression of bone regeneration. METHODS We histologically analyzed the spatio-temporal occurrence of cells of the innate immune system (macrophages), the adaptive immune system (B and T lymphocytes), and bone cells (osteoblasts and osteoclasts) in the fracture area of a femoral osteotomy over the healing time. This study was performed in a bone osteotomy gap mouse model. We also investigated two key challenges of successful bone regeneration: hypoxia and revascularization. RESULTS Macrophages were present in and around the fracture gap throughout the entire healing period. The switch from initially pro-inflammatory M1 macrophages to the anti-inflammatory M2 phenotype coincided with the revascularization as well as the appearance of osteoblasts in the fracture area. This indicates that M2 macrophages are necessary for the restoration of vessels and that they also play an orchestrating role in osteoblastogenesis during bone healing. The presence of adaptive immune cells throughout the healing process emphasizes their essential role for regenerative processes that exceeds a mere pathogen defense. B and T cells co-localize consistently with bone cells throughout the healing process, consolidating their crucial role in guiding bone formation. These histological data provide, for the first time, comprehensive information about the complex interrelationships of the cellular network during the entire bone healing process in one standardized set up. With this, an overall picture of the spatio-temporal interplay of cellular key players in a bone healing scenario has been created. CONCLUSIONS A spatio-temporal distribution of immune cells, bone cells, and factors driving bone healing at time points that are decisive for this process-especially during the initial steps of inflammation and revascularization, as well as the soft and hard callus phases-has been visualized. The results show that the bone healing cascade does not consist of five distinct, consecutive phases but is a rather complex interrelated and continuous process of events, especially at the onset of healing.
Collapse
Affiliation(s)
- Claudia Schlundt
- Julius Wolff Institut, BIH at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (C.S.); (R.A.S.); (C.H.B.); (G.N.D.)
- BIH Center for Regenerative Therapies, BIH at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany;
| | - Radost A. Saß
- Julius Wolff Institut, BIH at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (C.S.); (R.A.S.); (C.H.B.); (G.N.D.)
- BIH Center for Regenerative Therapies, BIH at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany;
| | - Christian H. Bucher
- Julius Wolff Institut, BIH at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (C.S.); (R.A.S.); (C.H.B.); (G.N.D.)
- BIH Center for Regenerative Therapies, BIH at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany;
| | - Sabine Bartosch
- Berlin School for Regenerative Therapies, Charité—Universitätsmedizin Berlin, Augustenburger Plarz 1, 13353 Berlin, Germany;
| | - Anja E. Hauser
- Rheumatology and Clinical Immunology, Charité—Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany;
- Immune Dynamics, Deutsches Rheuma-Forschungszentrum Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Hans-Dieter Volk
- BIH Center for Regenerative Therapies, BIH at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany;
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Georg N. Duda
- Julius Wolff Institut, BIH at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (C.S.); (R.A.S.); (C.H.B.); (G.N.D.)
- BIH Center for Regenerative Therapies, BIH at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany;
| | - Katharina Schmidt-Bleek
- Julius Wolff Institut, BIH at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (C.S.); (R.A.S.); (C.H.B.); (G.N.D.)
- BIH Center for Regenerative Therapies, BIH at Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany;
| |
Collapse
|
22
|
Mahajan A, Bhattacharyya S. Immunomodulation by mesenchymal stem cells during osteogenic differentiation: Clinical implications during bone regeneration. Mol Immunol 2023; 164:143-152. [PMID: 38011783 DOI: 10.1016/j.molimm.2023.11.006] [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: 06/12/2023] [Revised: 11/08/2023] [Accepted: 11/12/2023] [Indexed: 11/29/2023]
Abstract
Critical bone defects resulting in delayed and non-union are a major concern in the field of orthopedics. Over the past decade, mesenchymal stem cells (MSCs) have become a promising frontier for bone repair and regeneration owing to their high expansion rate and osteogenic differentiation potential ex vivo. MSCs have also long been associated with their ability to modulate immune response in the recipients. These can even skew the immune response towards pro-inflammatory or anti-inflammatory type by sensing their local microenvironment. MSCs adopt anti-inflammatory phenotype at bone injury site and secrete various immunomodulatory factors such as IDO, NO, TGFβ1 and PGE-2 which have redundant role in osteoblast differentiation and bone formation. As such, several studies have also sought to decipher the immunomodulatory effects of osteogenically differentiated MSCs. The present review discusses the immunomodulatory status of MSCs during their osteogenic differentiation and summarizes few mechanisms that cause immunosuppression by osteogenically differentiated MSCs and its implication during bone healing.
Collapse
Affiliation(s)
- Aditi Mahajan
- Department of Biophysics, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Shalmoli Bhattacharyya
- Department of Biophysics, Post Graduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
23
|
Borgiani E, Nasello G, Ory L, Herpelinck T, Groeneveldt L, Bucher CH, Schmidt-Bleek K, Geris L. COMMBINI: an experimentally-informed COmputational Model of Macrophage dynamics in the Bone INjury Immunoresponse. Front Immunol 2023; 14:1231329. [PMID: 38130715 PMCID: PMC10733790 DOI: 10.3389/fimmu.2023.1231329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 10/11/2023] [Indexed: 12/23/2023] Open
Abstract
Bone fracture healing is a well-orchestrated but complex process that involves numerous regulations at different scales. This complexity becomes particularly evident during the inflammatory stage, as immune cells invade the healing region and trigger a cascade of signals to promote a favorable regenerative environment. Thus, the emergence of criticalities during this stage might hinder the rest of the process. Therefore, the investigation of the many interactions that regulate the inflammation has a primary importance on the exploration of the overall healing progression. In this context, an in silico model named COMMBINI (COmputational Model of Macrophage dynamics in the Bone INjury Immunoresponse) has been developed to investigate the mechano-biological interactions during the early inflammatory stage at the tissue, cellular and molecular levels. An agent-based model is employed to simulate the behavior of immune cells, inflammatory cytokines and fracture debris as well as their reciprocal multiscale biological interactions during the development of the early inflammation (up to 5 days post-injury). The strength of the computational approach is the capacity of the in silico model to simulate the overall healing process by taking into account the numerous hidden events that contribute to its success. To calibrate the model, we present an in silico immunofluorescence method that enables a direct comparison at the cellular level between the model output and experimental immunofluorescent images. The combination of sensitivity analysis and a Genetic Algorithm allows dynamic cooperation between these techniques, enabling faster identification of the most accurate parameter values, reducing the disparity between computer simulation and histological data. The sensitivity analysis showed a higher sensibility of the computer model to the macrophage recruitment ratio during the early inflammation and to proliferation in the late stage. Furthermore, the Genetic Algorithm highlighted an underestimation of macrophage proliferation by in vitro experiments. Further experiments were conducted using another externally fixated murine model, providing an independent validation dataset. The validated COMMBINI platform serves as a novel tool to deepen the understanding of the intricacies of the early bone regeneration phases. COMMBINI aims to contribute to designing novel treatment strategies in both the biological and mechanical domains.
Collapse
Affiliation(s)
- Edoardo Borgiani
- Biomechanics Research Unit, GIGA-In Silico Medicine, University of Liège, Liège, Belgium
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Division of Biomechanics, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Gabriele Nasello
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Liesbeth Ory
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Tim Herpelinck
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Lisanne Groeneveldt
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Christian H. Bucher
- Julius Wolff Institute, Berlin Institute of Health, Charitè – Universitätsmedizin Berlin, Berlin, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute, Berlin Institute of Health, Charitè – Universitätsmedizin Berlin, Berlin, Germany
| | - Liesbet Geris
- Biomechanics Research Unit, GIGA-In Silico Medicine, University of Liège, Liège, Belgium
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
- Division of Biomechanics, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
- Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| |
Collapse
|
24
|
Heyland M, Deppe D, Reisener MJ, Damm P, Taylor WR, Reinke S, Duda GN, Trepczynski A. Lower-limb internal loading and potential consequences for fracture healing. Front Bioeng Biotechnol 2023; 11:1284091. [PMID: 37901836 PMCID: PMC10602681 DOI: 10.3389/fbioe.2023.1284091] [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: 08/27/2023] [Accepted: 10/02/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction: Mechanical loading is known to determine the course of bone fracture healing. We hypothesise that lower limb long bone loading differs with knee flexion angle during walking and frontal knee alignment, which affects fracture healing success. Materials and methods: Using our musculoskeletal in silico modelling constrained against in vivo data from patients with instrumented knee implants allowed us to assess internal loads in femur and tibia. These internal forces were associated with the clinical outcome of fracture healing in a relevant cohort of 178 extra-articular femur and tibia fractures in patients using a retrospective approach. Results: Mean peak forces differed with femoral compression (1,330-1,936 N at mid-shaft) amounting to about half of tibial compression (2,299-5,224 N). Mean peak bending moments in the frontal plane were greater in the femur (71-130 Nm) than in the tibia (from 26 to 43 Nm), each increasing proximally. Bending in the sagittal plane showed smaller mean peak bending moments in the femur (-38 to 43 Nm) reaching substantially higher values in the tibia (-63 to -175 Nm) with a peak proximally. Peak torsional moments had opposite directions for the femur (-13 to -40 Nm) versus tibia (15-48 Nm) with an increase towards the proximal end in both. Femoral fractures showed significantly lower scores in the modified Radiological Union Scale for Tibia (mRUST) at last follow-up (p < 0.001) compared to tibial fractures. Specifically, compression (r = 0.304), sagittal bending (r = 0.259), and frontal bending (r = -0.318) showed strong associations (p < 0.001) to mRUST at last follow-up. This was not the case for age, body weight, or localisation alone. Discussion: This study showed that moments in femur and tibia tend to decrease towards their distal ends. Tibial load components were influenced by knee flexion angle, especially at push-off, while static frontal alignment played a smaller role. Our results indicate that femur and tibia are loaded differently and thus require adapted fracture fixation considering load components rather than just overall load level.
Collapse
Affiliation(s)
- Mark Heyland
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Dominik Deppe
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Berlin, Germany
- Department of Radiology, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Marie Jacqueline Reisener
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Berlin, Germany
- Centre for Muskuloskeletal Surgery (CMSC), Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Philipp Damm
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - William R. Taylor
- Laboratory for Movement Biomechanics, ETH Zürich, Zürich, Switzerland
| | - Simon Reinke
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Georg N. Duda
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Adam Trepczynski
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
25
|
Mi B, Xiong Y, Zha K, Cao F, Zhou W, Abbaszadeh S, Ouyang L, Liao Y, Hu W, Dai G, Zhao Z, Feng Q, Shahbazi MA, Liu G. Immune homeostasis modulation by hydrogel-guided delivery systems: a tool for accelerated bone regeneration. Biomater Sci 2023; 11:6035-6059. [PMID: 37522328 DOI: 10.1039/d3bm00544e] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
Immune homeostasis is delicately mediated by the dynamic balance between effector immune cells and regulatory immune cells. Local deviations from immune homeostasis in the microenvironment of bone fractures, caused by an increased ratio of effector to regulatory cues, can lead to excessive inflammatory conditions and hinder bone regeneration. Therefore, achieving effective and localized immunomodulation of bone fractures is crucial for successful bone regeneration. Recent research has focused on developing localized and specific immunomodulatory strategies using local hydrogel-based delivery systems. In this review, we aim to emphasize the significant role of immune homeostasis in bone regeneration, explore local hydrogel-based delivery systems, discuss emerging trends in immunomodulation for enhancing bone regeneration, and address the limitations of current delivery strategies along with the challenges of clinical translation.
Collapse
Affiliation(s)
- Bobin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Yuan Xiong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Kangkang Zha
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Faqi Cao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Wu Zhou
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Samin Abbaszadeh
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Lizhi Ouyang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Yuheng Liao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Weixian Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Guandong Dai
- Department of Orthopedic Surgery, Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen 518118, China
| | - Zhiming Zhao
- Department of Orthopedics, Suizhou Hospital, Hubei University of Medicine, Suizhou 441300, China
| | - Qian Feng
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Mohammad-Ali Shahbazi
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | - Guohui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| |
Collapse
|
26
|
Söhling N, Heilani M, Fremdling C, Schaible A, Schröder K, Brune JC, Eras V, Nau C, Marzi I, Henrich D, Verboket RD. One Stage Masquelets Technique: Evaluation of Different Forms of Membrane Filling with and without Bone Marrow Mononuclear Cells (BMC) in Large Femoral Bone Defects in Rats. Cells 2023; 12:cells12091289. [PMID: 37174689 PMCID: PMC10177115 DOI: 10.3390/cells12091289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/23/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
The classic two-stage masquelet technique is an effective procedure for the treatment of large bone defects. Our group recently showed that one surgery could be saved by using a decellularized dermis membrane (DCD, Epiflex, DIZG). In addition, studies with bone substitute materials for defect filling show that it also appears possible to dispense with the removal of syngeneic cancellous bone (SCB), which is fraught with complications. The focus of this work was to clarify whether the SCB can be replaced by the granular demineralized bone matrix (g-DBM) or fibrous demineralized bone matrix (f-DBM) demineralized bone matrix and whether the colonization of the DCD and/or the DBM defect filling with bone marrow mononuclear cells (BMC) can lead to improved bone healing. In 100 Sprague Dawley rats, a critical femoral bone defect 5 mm in length was stabilized with a plate and then encased in DCD. Subsequently, the defect was filled with SCB (control), g-DBM, or f-DBM, with or without BMC. After 8 weeks, the femurs were harvested and subjected to histological, radiological, and biomechanical analysis. The analyses showed the incipient bony bridging of the defect zone in both groups for g-DBM and f-DBM. Stability and bone formation were not affected compared to the control group. The addition of BMCs showed no further improvement in bone healing. In conclusion, DBM offers a new perspective on defect filling; however, the addition of BMC did not lead to better results.
Collapse
Affiliation(s)
- Nicolas Söhling
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Myriam Heilani
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Charlotte Fremdling
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Alexander Schaible
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Katrin Schröder
- Center of Physiology, Cardiovascular Physiology, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Jan C Brune
- German Institute for Cell and Tissue Replacement (DIZG, gemeinnützige GmbH), 12555 Berlin, Germany
| | - Volker Eras
- German Institute for Cell and Tissue Replacement (DIZG, gemeinnützige GmbH), 12555 Berlin, Germany
| | - Christoph Nau
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Ingo Marzi
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Dirk Henrich
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - René D Verboket
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| |
Collapse
|
27
|
Dar HY, Perrien DS, Pal S, Stoica A, Uppuganti S, Nyman JS, Jones RM, Weitzmann MN, Pacifici R. Callus γδ T cells and microbe-induced intestinal Th17 cells improve fracture healing in mice. J Clin Invest 2023; 133:e166577. [PMID: 36881482 PMCID: PMC10104897 DOI: 10.1172/jci166577] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
IL-17A (IL-17), a driver of the inflammatory phase of fracture repair, is produced locally by several cell lineages including γδ T cells and Th17 cells. However, the origin of these T cells and their relevance for fracture repair are unknown. Here, we show that fractures rapidly expanded callus γδ T cells, which led to increased gut permeability by promoting systemic inflammation. When the microbiota contained the Th17 cell-inducing taxon segmented filamentous bacteria (SFB), activation of γδ T cells was followed by expansion of intestinal Th17 cells, their migration to the callus, and improved fracture repair. Mechanistically, fractures increased the S1P receptor 1-mediated (S1PR1-mediated) egress of Th17 cells from the intestine and enhanced their homing to the callus through a CCL20-mediated mechanism. Fracture repair was impaired by deletion of γδ T cells, depletion of the microbiome by antibiotics (Abx), blockade of Th17 cell egress from the gut, or Ab neutralization of Th17 cell influx into the callus. These findings demonstrate the relevance of the microbiome and T cell trafficking for fracture repair. Modifications of microbiome composition via Th17 cell-inducing bacteriotherapy and avoidance of broad-spectrum Abx may represent novel therapeutic strategies to optimize fracture healing.
Collapse
Affiliation(s)
- Hamid Y. Dar
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine and
- Emory Microbiome Research Center, Emory University, Atlanta, Georgia, USA
| | - Daniel S. Perrien
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine and
- Emory Microbiome Research Center, Emory University, Atlanta, Georgia, USA
| | - Subhashis Pal
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine and
- Emory Microbiome Research Center, Emory University, Atlanta, Georgia, USA
| | - Andreea Stoica
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine and
- Emory Microbiome Research Center, Emory University, Atlanta, Georgia, USA
| | - Sasidhar Uppuganti
- Department of Orthopedic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jeffry S. Nyman
- Department of Orthopedic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Rheinallt M. Jones
- Emory Microbiome Research Center, Emory University, Atlanta, Georgia, USA
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University, Atlanta, Georgia, USA
| | - M. Neale Weitzmann
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine and
- Emory Microbiome Research Center, Emory University, Atlanta, Georgia, USA
- Atlanta VA Health Care System, Department of Veterans Affairs, Decatur, Georgia, USA
| | - Roberto Pacifici
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine and
- Emory Microbiome Research Center, Emory University, Atlanta, Georgia, USA
- Immunology and Molecular Pathogenesis Program, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
28
|
Su N, Villicana C, Barati D, Freeman P, Luo Y, Yang F. Stem Cell Membrane-Coated Microribbon Scaffolds Induce Regenerative Innate and Adaptive Immune Responses in a Critical-Size Cranial Bone Defect Model. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2208781. [PMID: 36560890 PMCID: PMC10057912 DOI: 10.1002/adma.202208781] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/09/2022] [Indexed: 05/31/2023]
Abstract
Naturally-derived cell membranes have shown great promise in functionalizing nanoparticles to enhance biointerfacing functions for drug delivery applications. However, its potential for functionalizing macroporous scaffolds to enhance tissue regeneration in vivo remains unexplored. Engineering scaffolds with immunomodulatory functions represents an exciting strategy for tissue regeneration but is largely limited to soft tissues. Critical-sized bone defects cannot heal on their own, and the role of adaptive immune cells in scaffold-mediated healing of cranial bone defects remains largely unknown. Here, mensenchymal stem cell membrane (MSCM)-coated microribbon (µRB) scaffolds for treating critical size cranial bone defects via targeting immunomodulation are reported. Confocal imaging and proteomic analyses are used to confirm successful coating and characterize the compositions of cell membrane coating. It is demonstrated that MSCM coating promotes macrophage (Mφ) polarization toward regenerative phenotype, induces CD8+ T cell apoptosis, and enhances regulatory T cell differentiation in vitro and in vivo. When combined with a low dosage of BMP-2, MSCM coating further accelerates bone regeneration and suppresses inflammation. These results establish cell membrane-coated microribbon scaffolds as a promising strategy for treating critical size bone defects via immunomodulation. The platform may be broadly used with different cell membranes and scaffolds to enhance regeneration of multiple tissue types.
Collapse
Affiliation(s)
- Ni Su
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Cassandra Villicana
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Danial Barati
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Peyton Freeman
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Ying Luo
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA 02155
| | - Fan Yang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
29
|
McColl LF, Chen X, Solga MD, Schlegel K, Haughey SP, Lobo PI, Fread K, Zunder E, Cha R, Park S, Christophel JJ, Cui Q, Dighe AS. BMP-6 promotes type 2 immune response during enhancement of rat mandibular bone defect healing. Front Immunol 2023; 14:1064238. [PMID: 36845161 PMCID: PMC9950738 DOI: 10.3389/fimmu.2023.1064238] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 01/31/2023] [Indexed: 02/12/2023] Open
Abstract
Introduction Bone morphogenetic proteins (BMPs) are used as key therapeutic agents for the treatment of difficult fractures. While their effects on osteoprogenitors are known, little is known about their effects on the immune system. Methods We used permutations of BMP-6 (B), vascular endothelial growth factor (V), and Hedgehog signaling pathway activator smoothened agonist (S), to treat a rat mandibular defect and investigated healing outcomes at week 8, in correlation with the cellular landscape of the immune cells in the fracture callus at week 2. Results Maximum recruitment of immune cells to the fracture callus is known to occur at week 2. While the control, S, V, and VS groups remained as nonunions at week 8; all BMP-6 containing groups - B, BV, BS and BVS, showed near-complete to complete healing. This healing pattern was strongly associated with significantly higher ratios of CD4 T (CD45+CD3+CD4+) to putative CD8 T cells (CD45+CD3+CD4-), in groups treated with any permutation of BMP-6. Although, the numbers of putative M1 macrophages (CD45+CD3-CD11b/c+CD38high) were significantly lower in BMP-6 containing groups in comparison with S and VS groups, percentages of putative - Th1 cells or M1 macrophages (CD45+CD4+IFN-γ+) and putative - NK, NKT or cytotoxic CD8T cells (CD45+CD4-IFN-γ+) were similar in control and all treatment groups. Further interrogation revealed that the BMP-6 treatment promoted type 2 immune response by significantly increasing the numbers of CD45+CD3-CD11b/c+CD38low putative M2 macrophages, putative - Th2 cells or M2 macrophages (CD45+CD4+IL-4+) cells and putative - mast cells, eosinophils or basophils (CD45+CD4-IL-4+ cells). CD45- non-haematopoietic fractions of cells which encompass all known osteoprogenitor stem cells populations, were similar in control and treatment groups. Discussion This study uncovers previously unidentified regulatory functions of BMP-6 and shows that BMP-6 enhances fracture healing by not only acting on osteoprogenitor stem cells but also by promoting type 2 immune response.
Collapse
Affiliation(s)
- Logan F. McColl
- Department of Orthopedic Surgery, University of Virginia Health System, Charlottesville, VA, United States
| | - Xizhao Chen
- Department of Orthopedic Surgery, University of Virginia Health System, Charlottesville, VA, United States
| | - Michael D. Solga
- Flow Cytometry Core Facility, University of Virginia, Charlottesville, VA, United States
| | - Kailo Schlegel
- Department of Nephrology, University of Virginia Health System, Charlottesville, VA, United States
| | - Sean P. Haughey
- Department of Orthopedic Surgery, University of Virginia Health System, Charlottesville, VA, United States
| | - Peter I. Lobo
- Department of Nephrology, University of Virginia Health System, Charlottesville, VA, United States
| | - Kristen Fread
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Eli Zunder
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Ryan Cha
- Department of Orthopedic Surgery, University of Virginia Health System, Charlottesville, VA, United States
| | - Stephen Park
- Department of Otolaryngology–Head and Neck Surgery, University of Virginia Health System, Charlottesville, VA, United States
| | - J. Jared Christophel
- Department of Otolaryngology–Head and Neck Surgery, University of Virginia Health System, Charlottesville, VA, United States
| | - Quanjun Cui
- Department of Orthopedic Surgery, University of Virginia Health System, Charlottesville, VA, United States
| | - Abhijit S. Dighe
- Department of Orthopedic Surgery, University of Virginia Health System, Charlottesville, VA, United States,*Correspondence: Abhijit S. Dighe,
| |
Collapse
|
30
|
Haeusner S, Jauković A, Kupczyk E, Herrmann M. Review: cellularity in bone marrow autografts for bone and fracture healing. Am J Physiol Cell Physiol 2023; 324:C517-C531. [PMID: 36622067 DOI: 10.1152/ajpcell.00482.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The use of autografts, as primary cell and tissue source, is the current gold standard approach to treat critical size bone defects and nonunion defects. The unique mixture of the autografts, containing bony compartments and bone marrow (BM), delivers promising results. Although BM mesenchymal stromal cells (BM-MSCs) still represent a major target for various healing approaches in current preclinical research and respective clinical trials, their occurrence in the human BM is typically low. In vitro expansion of this cell type is regulatory challenging as well as time and cost intensive. Compared with marginal percentages of resident BM-MSCs in BM, BM mononuclear cells (BM-MNCs) contained in BM aspirates, concentrates, and bone autografts represent a readily available abundant cell source, applicable within hours during surgical procedures without the need for time-consuming and regulatory challenging cell expansion. This benefit is one reason why autografting has become a clinical standard procedure. However, the exact anatomy and cellularity of BM-MNCs in humans, which is strongly correlated to their unique mode of action and wide application range remains to be elucidated. The aim of this review was to present an overview of the current knowledge on these specific cell types found in human BM, emphasize the contribution of BM-MNCs in bone healing, highlight donor site dependence, and discuss limitations in the current isolation and subsequent characterization procedures. Hereby, the most recent and relevant examples of human BM-MNC cell characterization, flow cytometric analyses, and findings are summarized, with a strong focus on bone therapy.
Collapse
Affiliation(s)
- S Haeusner
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital of Wuerzburg, Wuerzburg, Germany.,Bernhard-Heine-Center for Locomotion Research, University of Wuerzburg, Wuerzburg, Germany
| | - A Jauković
- Group for Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - E Kupczyk
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - M Herrmann
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital of Wuerzburg, Wuerzburg, Germany.,Bernhard-Heine-Center for Locomotion Research, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
31
|
Duda GN, Geissler S, Checa S, Tsitsilonis S, Petersen A, Schmidt-Bleek K. The decisive early phase of bone regeneration. Nat Rev Rheumatol 2023; 19:78-95. [PMID: 36624263 DOI: 10.1038/s41584-022-00887-0] [Citation(s) in RCA: 125] [Impact Index Per Article: 62.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2022] [Indexed: 01/11/2023]
Abstract
Bone has a remarkable endogenous regenerative capacity that enables scarless healing and restoration of its prior mechanical function, even under challenging conditions such as advanced age and metabolic or immunological degenerative diseases. However - despite much progress - a high number of bone injuries still heal with unsatisfactory outcomes. The mechanisms leading to impaired healing are heterogeneous, and involve exuberant and non-resolving immune reactions or overstrained mechanical conditions that affect the delicate regulation of the early initiation of scar-free healing. Every healing process begins phylogenetically with an inflammatory reaction, but its spatial and temporal intensity must be tightly controlled. Dysregulation of this inflammatory cascade directly affects the subsequent healing phases and hinders the healing progression. This Review discusses the complex processes underlying bone regeneration, focusing on the early healing phase and its highly dynamic environment, where vibrant changes in cellular and tissue composition alter the mechanical environment and thus affect the signalling pathways that orchestrate the healing process. Essential to scar-free healing is the interplay of various dynamic cascades that control timely resolution of local inflammation and tissue self-organization, while also providing sufficient local stability to initiate endogenous restoration. Various immunotherapy and mechanobiology-based therapy options are under investigation for promoting bone regeneration.
Collapse
Affiliation(s)
- Georg N Duda
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany. .,Berlin Institute of Health Centre for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.
| | - Sven Geissler
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health Centre for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sara Checa
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Serafeim Tsitsilonis
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health Centre for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ansgar Petersen
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health Centre for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health Centre for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
32
|
Hatt LP, Armiento AR, Mys K, Thompson K, Hildebrand M, Nehrbass D, Müller WEG, Zeiter S, Eglin D, Stoddart MJ. Standard in vitro evaluations of engineered bone substitutes are not sufficient to predict in vivo preclinical model outcomes. Acta Biomater 2023; 156:177-189. [PMID: 35988660 DOI: 10.1016/j.actbio.2022.08.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 01/18/2023]
Abstract
Understanding the optimal conditions required for bone healing can have a substantial impact to target the problem of non-unions and large bone defects. The combination of bioactive factors, regenerative progenitor cells and biomaterials to form a tissue engineered (TE) complex is a promising solution but translation to the clinic has been slow. We hypothesized the typical material testing algorithm used is insufficient and leads to materials being mischaracterized as promising. In the first part of this study, human bone marrow - derived mesenchymal stromal cells (hBM-MSCs) were embedded in three commonly used biomaterials (hyaluronic acid methacrylate, gelatin methacrylate and fibrin) and combined with relevant bioactive osteogenesis factors (dexamethasone microparticles and polyphosphate nanoparticles) to form a TE construct that underwent in vitro osteogenic differentiation for 28 days. Gene expression of relevant transcription factors and osteogenic markers, and von Kossa staining were performed. In the second and third part of this study, the same combination of TE constructs were implanted subcutaneously (cell containing) in T cell-deficient athymic Crl:NIH-Foxn1rnu rats for 8 weeks or cell free in an immunocompetent New Zealand white rabbit calvarial model for 6 weeks, respectively. Osteogenic performance was investigated via MicroCT imaging and histology staining. The in vitro study showed enhanced upregulation of relevant genes and significant mineral deposition within the three biomaterials, generally considered as a positive result. Subcutaneous implantation indicates none to minor ectopic bone formation. No enhanced calvarial bone healing was detected in implanted biomaterials compared to the empty defect. The reasons for the poor correlation of in vitro and in vivo outcomes are unclear and needs further investigation. This study highlights the discrepancy between in vitro and in vivo outcomes, demonstrating that in vitro data should be interpreted with extreme caution. In vitro models with higher complexity are necessary to increase value for translational studies. STATEMENT OF SIGNIFICANCE: Preclinical testing of newly developed biomaterials is a crucial element of the development cycle. Despite this, there is still significant discrepancy between in vitro and in vivo test results. Within this study we investigate multiple combinations of materials and osteogenic stimulants and demonstrate a poor correlation between the in vitro and in vivo data. We propose rationale for why this may be the case and suggest a modified testing algorithm.
Collapse
Affiliation(s)
- Luan P Hatt
- AO Research Institute Davos, 7270 Davos Platz, Switzerland; Institute for Biomechanics, ETH Zürich; 8093 Zürich, Switzerland
| | | | - Karen Mys
- AO Research Institute Davos, 7270 Davos Platz, Switzerland
| | - Keith Thompson
- AO Research Institute Davos, 7270 Davos Platz, Switzerland
| | | | - Dirk Nehrbass
- AO Research Institute Davos, 7270 Davos Platz, Switzerland
| | - Werner E G Müller
- Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Stephan Zeiter
- AO Research Institute Davos, 7270 Davos Platz, Switzerland
| | - David Eglin
- Mines Saint-Etienne, Univ Lyon, Univ Jean Monnet, INSERM, U 1059 Sainbiose, Centre CIS, F-42023 Saint-Etienne, France
| | | |
Collapse
|
33
|
Burkhardt LM, Bucher CH, Löffler J, Rinne C, Duda GN, Geissler S, Schulz TJ, Schmidt-Bleek K. The benefits of adipocyte metabolism in bone health and regeneration. Front Cell Dev Biol 2023; 11:1104709. [PMID: 36895792 PMCID: PMC9988968 DOI: 10.3389/fcell.2023.1104709] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
Patients suffering from musculoskeletal diseases must cope with a diminished quality of life and an increased burden on medical expenses. The interaction of immune cells and mesenchymal stromal cells during bone regeneration is one of the key requirements for the restoration of skeletal integrity. While stromal cells of the osteo-chondral lineage support bone regeneration, an excessive accumulation of cells of the adipogenic lineage is thought to promote low-grade inflammation and impair bone regeneration. Increasing evidence indicates that pro-inflammatory signaling from adipocytes is responsible for various chronic musculoskeletal diseases. This review aims to summarize the features of bone marrow adipocytes by phenotype, function, secretory features, metabolic properties and their impact on bone formation. In detail, the master regulator of adipogenesis and prominent diabetes drug target, peroxisome proliferator-activated receptor γ (PPARG), will be debated as a potential therapeutic approach to enhance bone regeneration. We will explore the possibilities of using clinically established PPARG agonists, the thiazolidinediones (TZDs), as a treatment strategy to guide the induction of a pro-regenerative, metabolically active bone marrow adipose tissue. The impact of this PPARG induced bone marrow adipose tissue type on providing the necessary metabolites to sustain osteogenic-as well as beneficial immune cells during bone fracture healing will be highlighted.
Collapse
Affiliation(s)
- Lisa-Marie Burkhardt
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Christian H Bucher
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Julia Löffler
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Charlotte Rinne
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition, Potsdam-Rehbrücke, Nuthetal, Germany
| | - Georg N Duda
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Sven Geissler
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Tim J Schulz
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition, Potsdam-Rehbrücke, Nuthetal, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany.,University of Potsdam, Institute of Nutritional Science, Nuthetal, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute, Berlin Institute of Health (BIH) Charité, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| |
Collapse
|
34
|
Ren Y, Bäcker H, Müller M, Kienzle A. The role of myeloid derived suppressor cells in musculoskeletal disorders. Front Immunol 2023; 14:1139683. [PMID: 36936946 PMCID: PMC10020351 DOI: 10.3389/fimmu.2023.1139683] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
The immune system is closely linked to bone homeostasis and plays a pivotal role in several pathological and inflammatory conditions. Through various pathways it modulates various bone cells and subsequently sustains the physiological bone metabolism. Myeloid-derived suppressor cells (MDSCs) are a group of heterogeneous immature myeloid-derived cells that can exert an immunosuppressive function through a direct cell-to-cell contact, secretion of anti-inflammatory cytokines or specific exosomes. These cells mediate the innate immune response to chronic stress on the skeletal system. In chronic inflammation, MDSCs act as an inner offset to rebalance overactivation of the immune system. Moreover, they have been found to be involved in processes responsible for bone remodeling in different musculoskeletal disorders, autoimmune diseases, infection, and cancer. These cells can not only cause bone erosion by differentiating into osteoclasts, but also alleviate the immune reaction, subsequently leading to long-lastingly impacted bone remodeling. In this review, we discuss the impact of MDSCs on the bone metabolism under several pathological conditions, the involved modulatory pathways as well as potential therapeutic targets in MDSCs to improve bone health.
Collapse
Affiliation(s)
- Yi Ren
- Center for Musculoskeletal Surgery, Clinic for Orthopedics, Charité University Hospital, Berlin, Germany
| | - Henrik Bäcker
- Department of Orthopedics, Auckland City Hospital, Auckland, New Zealand
| | - Michael Müller
- Center for Musculoskeletal Surgery, Clinic for Orthopedics, Charité University Hospital, Berlin, Germany
| | - Arne Kienzle
- Center for Musculoskeletal Surgery, Clinic for Orthopedics, Charité University Hospital, Berlin, Germany
- BIH Charité Clinician Scientist Program, BIH Biomedical Innovation Academy, Berlin Institute of Health, Charité — Universitätsmedizin Berlin, Berlin, Germany
- *Correspondence: Arne Kienzle,
| |
Collapse
|
35
|
Lopas LA, Shen H, Zhang N, Jang Y, Tawfik VL, Goodman SB, Natoli RM. Clinical Assessments of Fracture Healing and Basic Science Correlates: Is There Room for Convergence? Curr Osteoporos Rep 2022; 21:216-227. [PMID: 36534307 DOI: 10.1007/s11914-022-00770-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/11/2022] [Indexed: 12/23/2022]
Abstract
PURPOSE OF REVIEW The purpose of this review is to summarize the clinical and basic science methods used to assess fracture healing and propose a framework to improve the translational possibilities. RECENT FINDINGS Mainstays of fracture healing assessment include clinical examination, various imaging modalities, and assessment of function. Pre-clinical studies have yielded insight into biomechanical progression as well as the genetic, molecular, and cellular processes of fracture healing. Efforts are emerging to identify early markers to predict impaired healing and possibly early intervention to alter these processes. Despite of the differences in clinical and preclinical research, opportunities exist to unify and improve the translational efforts between these arenas to develop and optimize our ability to assess and predict fracture healing, thereby improving the clinical care of these patients.
Collapse
Affiliation(s)
- Luke A Lopas
- Department of Orthopaedic Surgery, Indiana University School of Medicine, 1801 N. Senate Blvd Suite 535, Indianapolis, IN, USA.
| | - Huaishuang Shen
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Orthopaedic Surgery, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Yohan Jang
- Department of Orthopaedic Surgery, Indiana University School of Medicine, 1801 N. Senate Blvd Suite 535, Indianapolis, IN, USA
| | - Vivianne L Tawfik
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Roman M Natoli
- Department of Orthopaedic Surgery, Indiana University School of Medicine, 1801 N. Senate Blvd Suite 535, Indianapolis, IN, USA
| |
Collapse
|
36
|
Shirazi S, Ravindran S, Cooper LF. Topography-mediated immunomodulation in osseointegration; Ally or Enemy. Biomaterials 2022; 291:121903. [PMID: 36410109 PMCID: PMC10148651 DOI: 10.1016/j.biomaterials.2022.121903] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022]
Abstract
Osteoimmunology is at full display during endosseous implant osseointegration. Bone formation, maintenance and resorption at the implant surface is a result of bidirectional and dynamic reciprocal communication between the bone and immune cells that extends beyond the well-defined osteoblast-osteoclast signaling. Implant surface topography informs adherent progenitor and immune cell function and their cross-talk to modulate the process of bone accrual. Integrating titanium surface engineering with the principles of immunology is utilized to harness the power of immune system to improve osseointegration in healthy and diseased microenvironments. This review summarizes current information regarding immune cell-titanium implant surface interactions and places these events in the context of surface-mediated immunomodulation and bone regeneration. A mechanistic approach is directed in demonstrating the central role of osteoimmunology in the process of osseointegration and exploring how regulation of immune cell function at the implant-bone interface may be used in future control of clinical therapies. The process of peri-implant bone loss is also informed by immunomodulation at the implant surface. How surface topography is exploited to prevent osteoclastogenesis is considered herein with respect to peri-implant inflammation, osteoclastic precursor-surface interactions, and the upstream/downstream effects of surface topography on immune and progenitor cell function.
Collapse
Affiliation(s)
- Sajjad Shirazi
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL, USA.
| | - Sriram Ravindran
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL, USA
| | - Lyndon F Cooper
- School of Dentistry, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
37
|
Kang IH, Baliga UK, Chatterjee S, Chakraborty P, Choi S, Buchweitz N, Li H, Wu Y, Yao H, Mehrotra S, Mehrotra M. Quantitative increase in T regulatory cells enhances bone remodeling in osteogenesis imperfecta. iScience 2022; 25:104818. [PMID: 36034228 PMCID: PMC9400089 DOI: 10.1016/j.isci.2022.104818] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 06/07/2022] [Accepted: 07/19/2022] [Indexed: 02/03/2023] Open
Abstract
Osteogenesis imperfecta (OI) is characterized by repeated bone fractures. Recent studies have shown that T lymphocytes and regulatory T cells (Tregs) regulate the functions of osteoclasts and osteoblasts, thus playing a role in bone turnover. We demonstrate an activated effector phenotype and higher secretion of pro-inflammatory cytokines, IFN-γ, and TNF-α in OI peripheral T cells as compared with wild-type (WT). Suppressive Tregs (spleen and thymus) were qualitatively similar, whereas there was a quantitative decrease in OI versus WT. Restoring Treg numbers by systemic transplantation in OI mice resulted in reduced T cell activation and effector cytokine secretion that correlated with significant improvements in tibial trabecular and cortical bone parameters and stiffness of femur, along with increased osteoblast mineralization and decreased osteoclast numbers. Therefore, Tregs can dampen the pro-inflammatory environment and enhance bone remodeling in OI mice. Thus, this study will be helpful in developing future autologous immunotherapy-based treatment modalities for OI.
Collapse
Affiliation(s)
- In-Hong Kang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Uday K. Baliga
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Shilpak Chatterjee
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Paramita Chakraborty
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Seungho Choi
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Nathan Buchweitz
- Department of Orthopedics, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Bioengineering, Clemson University, Clemson, SC 29634, USA
- Clemson-MUSC Joint Bioengineering Program, South Carolina, USA
| | - Hong Li
- Depatment of Public Health Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Yongren Wu
- Department of Orthopedics, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Bioengineering, Clemson University, Clemson, SC 29634, USA
- Clemson-MUSC Joint Bioengineering Program, South Carolina, USA
| | - Hai Yao
- Department of Orthopedics, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Bioengineering, Clemson University, Clemson, SC 29634, USA
- Clemson-MUSC Joint Bioengineering Program, South Carolina, USA
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Meenal Mehrotra
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
- Center for Oral Health Research, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
38
|
Chow SKH, Wong CHW, Cui C, Li MMC, Wong RMY, Cheung WH. Modulating macrophage polarization for the enhancement of fracture healing, a systematic review. J Orthop Translat 2022; 36:83-90. [PMID: 35979176 PMCID: PMC9364046 DOI: 10.1016/j.jot.2022.05.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 12/04/2022] Open
Abstract
Background All fracture repairs start with the innate immune system with the inflammatory response known as the inflammatory stage guided and driven by the secretion of chemokine by the ruptured tissue, followed by the sequential recruitment of neutrophils, monocytes and macrophages. These innate immune cells would infiltrate the fracture site and secrete inflammatory cytokines to stimulate recruitment of more immune cells to arrive at the fracture site coordinating subsequent stages of the repair process. In which, subsidence of pro-inflammatory M1 macrophage and transformation to anti-inflammatory M2 macrophages promotes osteogenesis that marks the start of the anabolic endochondral stage. Methods Literature search was performed on Pubmed, Embase, and Web of Science databases (last accessed 15th April 2021) using “macrophage AND fracture”. Review was performed in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guideline. Results Eleven pre-clinical animal studies out of 429 articles were included in this systematic review according to our inclusion and exclusion criteria. All of which investigated interventions targeting to modulate the acute inflammatory response and macrophage polarization as evident by various markers in association with fracture healing outcomes. Conclusion This systematic review summarizes attempts to modulate the innate immune response with focuses on promoting macrophage polarization from M1 to M2 phenotype targeting the enhancement of fracture injury repair. Methods used to achieve the goal may include applications of damage-associated molecular pattern (DAMP), pathogen-associated molecular pattern (PAMP) or mechanical stimulation that hold high translational potentials for clinical application in the near future.
Collapse
Affiliation(s)
- Simon Kwoon-Ho Chow
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Carissa Hing-Wai Wong
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Can Cui
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Michelle Meng-Chen Li
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ronald Man Yeung Wong
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wing-Hoi Cheung
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
39
|
Local immune cell contributions to fracture healing in aged individuals - A novel role for interleukin 22. EXPERIMENTAL & MOLECULAR MEDICINE 2022; 54:1262-1276. [PMID: 36028760 PMCID: PMC9440089 DOI: 10.1038/s12276-022-00834-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 04/25/2022] [Accepted: 06/06/2022] [Indexed: 11/08/2022]
Abstract
With increasing age, the risk of bone fractures increases while regenerative capacity decreases. This variation in healing potential appears to be linked to adaptive immunity, but the underlying mechanism is still unknown. This study sheds light on immunoaging/inflammaging, which impacts regenerative processes in aging individuals. In an aged preclinical model system, different levels of immunoaging were analyzed to identify key factors that connect immunoaged/inflammaged conditions with bone formation after long bone fracture. Immunological facets, progenitor cells, the microbiome, and confounders were monitored locally at the injury site and systemically in relation to healing outcomes in 12-month-old mice with distinct individual levels of immunoaging. Bone tissue formation during healing was delayed in the immunoaged group and could be associated with significant changes in cytokine levels. A prolonged and amplified pro-inflammatory reaction was caused by upregulated immune cell activation markers, increased chemokine receptor availability and a lack of inhibitory signaling. In immunoaged mice, interleukin-22 was identified as a core cell signaling protein that played a central role in delayed healing. Therapeutic neutralization of IL-22 reversed this specific immunoaging-related disturbed healing. Immunoaging was found to be an influencing factor of decreased regenerative capacity in aged individuals. Furthermore, a novel therapeutic strategy of neutralizing IL-22 may successfully rejuvenate healing in individuals with advanced immune experiences.
Collapse
|
40
|
Su N, Villicana C, Yang F. Immunomodulatory strategies for bone regeneration: A review from the perspective of disease types. Biomaterials 2022; 286:121604. [PMID: 35667249 PMCID: PMC9881498 DOI: 10.1016/j.biomaterials.2022.121604] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/19/2022] [Accepted: 05/24/2022] [Indexed: 02/08/2023]
Abstract
Tissue engineering strategies for treating bone loss to date have largely focused on targeting stem cells or vascularization. Immune cells, including macrophages and T cells, can also indirectly enhance bone healing via cytokine secretion to interact with other bone niche cells. Bone niche cues and local immune environment vary depending on anatomical location, size of defects and disease types. As such, it is critical to evaluate the role of the immune system in the context of specific bone niche and different disease types. This review focuses on immunomodulation research for bone applications using biomaterials and cell-based strategies, with a unique perspective from different disease types. We first reviewed applications for prolonging orthopaedic implant lifetime and enhancing fracture healing, two clinical challenges where immunomodulatory strategies were initially developed for orthopedic applications. We then reviewed recent research progress in harnessing immunomodulatory strategies for regenerating critical-sized, long bone or cranial bone defects, and treating osteolytic bone diseases. Remaining gaps in knowledge, future directions and opportunities were also discussed.
Collapse
Affiliation(s)
- Ni Su
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Cassandra Villicana
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Fan Yang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA.,Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, 94305, USA.,: Corresponding Author Fan Yang, Ph D, Department of Orthopaedic Surgery and Bioengineering, Stanford University School of Medicine, 240 Pasteur Dr, Palo Alto, CA 94304, Biomedical Innovation Building, 1st floor, Room 1200, , Phone: (650) 646-8558
| |
Collapse
|
41
|
Polikarpova A, Ellinghaus A, Schmidt-Bleek O, Grosser L, Bucher CH, Duda GN, Tanaka EM, Schmidt-Bleek K. The specialist in regeneration-the Axolotl-a suitable model to study bone healing? NPJ Regen Med 2022; 7:35. [PMID: 35773262 PMCID: PMC9246919 DOI: 10.1038/s41536-022-00229-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 05/31/2022] [Indexed: 11/08/2022] Open
Abstract
While the axolotl's ability to completely regenerate amputated limbs is well known and studied, the mechanism of axolotl bone fracture healing remains poorly understood. One reason might be the lack of a standardized fracture fixation in axolotl. We present a surgical technique to stabilize the osteotomized axolotl femur with a fixator plate and compare it to a non-stabilized osteotomy and to limb amputation. The healing outcome was evaluated 3 weeks, 3, 6 and 9 months post-surgery by microcomputer tomography, histology and immunohistochemistry. Plate-fixated femurs regained bone integrity more efficiently in comparison to the non-fixated osteotomized bone, where larger callus formed, possibly to compensate for the bone fragment misalignment. The healing of a non-critical osteotomy in axolotl was incomplete after 9 months, while amputated limbs efficiently restored bone length and structure. In axolotl amputated limbs, plate-fixated and non-fixated fractures, we observed accumulation of PCNA+ proliferating cells at 3 weeks post-injury similar to mouse. Additionally, as in mouse, SOX9-expressing cells appeared in the early phase of fracture healing and amputated limb regeneration in axolotl, preceding cartilage formation. This implicates endochondral ossification to be the probable mechanism of bone healing in axolotls. Altogether, the surgery with a standardized fixation technique demonstrated here allows for controlled axolotl bone healing experiments, facilitating their comparison to mammals (mice).
Collapse
Affiliation(s)
- A Polikarpova
- Research Institute of Molecular Pathology, Vienna, A-1030, Austria
| | - A Ellinghaus
- Julius Wolff Institute and BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, DE-13353, Germany
| | - O Schmidt-Bleek
- Julius Wolff Institute and BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, DE-13353, Germany
| | - L Grosser
- Research Institute of Molecular Pathology, Vienna, A-1030, Austria
| | - C H Bucher
- Julius Wolff Institute and BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, DE-13353, Germany
| | - G N Duda
- Julius Wolff Institute and BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, DE-13353, Germany
| | - E M Tanaka
- Research Institute of Molecular Pathology, Vienna, A-1030, Austria
| | - K Schmidt-Bleek
- Julius Wolff Institute and BIH Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, DE-13353, Germany.
| |
Collapse
|
42
|
Shang L, Shao J, Ge S. Immunomodulatory Properties: The Accelerant of Hydroxyapatite-Based Materials for Bone Regeneration. Tissue Eng Part C Methods 2022; 28:377-392. [PMID: 35196904 DOI: 10.1089/ten.tec.2022.00111112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The immunoinflammatory response is the prerequisite step for wound healing and tissue regeneration, and the immunomodulatory effects of biomaterials have attracted increasing attention. Hydroxyapatite [Ca10(PO4)6(OH)2] (HAp), a common calcium phosphate ceramic, due to its structural and functional similarity to the inorganic constituent of natural bones, has been developed for different application purposes such as bone substitutes, tissue engineering scaffolds, and implant coatings. Recently, the interaction between HAp-based materials and the immune system (various immune cells), and the immunomodulatory effects of HAp-based materials on bone tissue regeneration have been explored extensively. Macrophages-mediated regenerative effect by HAp stimulation occupies the mainstream status of immunomodulatory strategies. The immunomodulation of HAp can be manipulated by tuning the physical, chemical, and biological cues such as surface functionalization (physical or chemical modifications), structural and textural characteristics (size, shape, and surface topography), and the incorporation of bioactive substances (cytokines, rare-earth elements, and bioactive ions). Therefore, HAp ceramic materials can contribute to bone regeneration by creating a favorable osteoimmune microenvironment, which would provide a more comprehensive theoretical basis for their further clinical applications. Considering the rapidly developed HAp-based materials as well as their excellent biological performances in the field of regenerative medicine, this review discusses the recent advances concerning the immunomodulatory methods for HAp-based biomaterials and their roles in bone tissue regeneration.
Collapse
Affiliation(s)
- Lingling Shang
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Jinlong Shao
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Shaohua Ge
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| |
Collapse
|
43
|
Mi B, Chen L, Xiong Y, Yang Y, Panayi AC, Xue H, Hu Y, Yan C, Hu L, Xie X, Lin Z, Zhou W, Cao F, Xiao X, Feng Q, Liu G. Osteoblast/Osteoclast and Immune Cocktail Therapy of an Exosome/Drug Delivery Multifunctional Hydrogel Accelerates Fracture Repair. ACS NANO 2022; 16:771-782. [PMID: 34979087 DOI: 10.1021/acsnano.1c08284] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The osteoblast/osteoclast and M1/M2 macrophage ratios play critical roles in delayed fracture healing. Robust osteoblast differentiation and M2 macrophage polarization can substantiality promote fracture repair; however, the combined effect of these strategies has not been previously studied. In this study, we constructed a cocktail therapy to simultaneously regulate the osteoblast/osteoclast and M1/M2 macrophage balance. The cocktail therapy composed of a natural polymer hyaluronic-acid-based hydrogel (HA hydrogel, which has a tissue-adhesive, injectable, self-healing, anti-inflammation profile), engineered endothelial cell-derived exosomes (EC-ExosmiR-26a-5p), and APY29, an IRE-1α inhibitor. This allowed for specific delivery of EC-ExosmiR-26a-5p and APY29 for osteoblast/osteoclast and macrophage regulation, respectively. The results suggested that the cocktail therapy exerted pro-fracture repair effects with each of its components established as indispensable. The assessed cocktail therapy provides insight into synergistic strategies and is useful for developing more suitable pro-fracture repair therapy.
Collapse
Affiliation(s)
- Bobin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Lang Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Yuan Xiong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Yayan Yang
- College of Chemistry and Materials Science, Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian Normal University, Fuzhou 350007, China
| | - Adriana C Panayi
- Department of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School Boston, Massachusetts 02152, United States
| | - Hang Xue
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Yiqiang Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Chenchen Yan
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Liangcong Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Xudong Xie
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Ze Lin
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Wu Zhou
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Faqi Cao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Xiufeng Xiao
- College of Chemistry and Materials Science, Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian Normal University, Fuzhou 350007, China
| | - Qian Feng
- College of Chemistry and Materials Science, Fujian Provincial Key Laboratory of Advanced Materials Oriented Chemical Engineering, Fujian Normal University, Fuzhou 350007, China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Guohui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| |
Collapse
|
44
|
Cui Y, Li H, Li Y, Mao L. Novel insights into nanomaterials for immunomodulatory bone regeneration. NANOSCALE ADVANCES 2022; 4:334-352. [PMID: 36132687 PMCID: PMC9418834 DOI: 10.1039/d1na00741f] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 11/13/2021] [Indexed: 05/02/2023]
Abstract
Bone defect repair caused by trauma, congenital malformation, tumors, infection or systemic diseases remains the focus of attention in regeneration medicine. Recent advances in osteoimmunology indicate that immune cells and correlative cytokines modulate the delicate balance between osteoblasts and osteoclasts and induce a favorable microenvironment for bone regeneration. With superior attributes that imitate the three-dimensional architecture of natural bone, excellent fabricability, mechanical and biological properties, nanomaterials (NMs) are becoming attractive in the field of bone tissue engineering. Particularly, it could be an effective strategy for immunomodulatory bone regeneration by engineering NMs involved in composition nature, nanoarchitectural morphology, surface chemistry, topography and biological molecules, whose mechanisms potentially refer to regulating the phenotype of high-plastic immune cells and inducing cytokine secretion to accelerate osteogenesis. Despite these prominent achievements, the employment of NMs is poorly translated into clinical trials due to the lack of knowledge about the interaction between NMs and the immune system. For this reason, we sketch out the hierarchical structure of bone and its natural healing process, followed by discussion about the effects of immune cells on bone regeneration. Novel horizons focusing on recent progressions in the architectural and physicochemical performances of NMs and their impacts on the body defence mechanism are also emphasized, hoping to provide novel insights for the fabrication of bone graft materials in tissue engineering.
Collapse
Affiliation(s)
- Ya Cui
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology Shanghai China
| | - Hairui Li
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology Shanghai China
| | - Yaxin Li
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology Shanghai China
| | - Lixia Mao
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology Shanghai China
| |
Collapse
|
45
|
Ehnert S, Relja B, Schmidt-Bleek K, Fischer V, Ignatius A, Linnemann C, Rinderknecht H, Huber-Lang M, Kalbitz M, Histing T, Nussler AK. Effects of immune cells on mesenchymal stem cells during fracture healing. World J Stem Cells 2021; 13:1667-1695. [PMID: 34909117 PMCID: PMC8641016 DOI: 10.4252/wjsc.v13.i11.1667] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/31/2021] [Accepted: 09/03/2021] [Indexed: 02/06/2023] Open
Abstract
In vertebrates, bone is considered an osteoimmune system which encompasses functions of a locomotive organ, a mineral reservoir, a hormonal organ, a stem cell pool and a cradle for immune cells. This osteoimmune system is based on cooperatively acting bone and immune cells, cohabitating within the bone marrow. They are highly interdependent, a fact that is confounded by shared progenitors, mediators, and signaling pathways. Successful fracture healing requires the participation of all the precursors, immune and bone cells found in the osteoimmune system. Recent evidence demonstrated that changes of the immune cell composition and function may negatively influence bone healing. In this review, first the interplay between different immune cell types and osteoprogenitor cells will be elaborated more closely. The separate paragraphs focus on the specific cell types, starting with the cells of the innate immune response followed by cells of the adaptive immune response, and the complement system as mediator between them. Finally, a brief overview on the challenges of preclinical testing of immune-based therapeutic strategies to support fracture healing will be given.
Collapse
Affiliation(s)
- Sabrina Ehnert
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Tübingen 72076, Germany
| | - Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke University, Magdeburg 39120, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute and Berlin Institute of Health Center of Regenerative Therapies, Charité - University Medicine Berlin, Berlin 13353, Germany
| | - Verena Fischer
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm 89091, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm 89091, Germany
| | - Caren Linnemann
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Tübingen 72076, Germany
| | - Helen Rinderknecht
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Tübingen 72076, Germany
| | - Markus Huber-Lang
- Institute for Clinical and Experimental Trauma-Immunology (ITI), University Hospital Ulm, Ulm 89091, Germany
| | - Miriam Kalbitz
- Department of Trauma and Orthopedic Surgery, University Hospital Erlangen Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen 91054, Germany
| | - Tina Histing
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Tübingen 72076, Germany
| | - Andreas K Nussler
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Tübingen 72076, Germany
| |
Collapse
|
46
|
Turajane K, Ji G, Chinenov Y, Chao M, Ayturk U, Suhardi VJ, Greenblatt MB, Ivashkiv LB, Bostrom MPG, Yang X. RNA-seq Analysis of Peri-Implant Tissue Shows Differences in Immune, Notch, Wnt, and Angiogenesis Pathways in Aged Versus Young Mice. JBMR Plus 2021; 5:e10535. [PMID: 34761143 PMCID: PMC8567488 DOI: 10.1002/jbm4.10535] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/19/2021] [Accepted: 07/27/2021] [Indexed: 12/16/2022] Open
Abstract
The number of total joint replacements (TJRs) in the United States is increasing annually. Cementless implants are intended to improve upon traditional cemented implants by allowing bone growth directly on the surface to improve implant longevity. One major complication of TJR is implant loosening, which is related to deficient osseointegration in cementless TJRs. Although poor osseointegration in aged patients is typically attributed to decreased basal bone mass, little is known about the molecular pathways that compromise the growth of bone onto porous titanium implants. To identify the pathways important for osseointegration that are compromised by aging, we developed an approach for transcriptomic profiling of peri-implant tissue in young and aged mice using our murine model of osseointegration. Based on previous findings of changes of bone quality associated with aging, we hypothesized that aged mice have impaired activation of bone anabolic pathways at the bone-implant interface. We found that pathways most significantly downregulated in aged mice relative to young mice are related to angiogenic, Notch, and Wnt signaling. Downregulation of these pathways is associated with markedly increased expression of inflammatory and immune genes at the bone-implant interface in aged mice. These results identify osseointegration pathways affected by aging and suggest that an increased inflammatory response in aged mice may compromise peri-implant bone healing. Targeting the Notch and Wnt pathways, promoting angiogenesis, or modulating the immune response at the peri-implant site may enhance osseointegration and improve the outcome of joint replacement in older patients. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
| | - Gang Ji
- Hospital for Special SurgeryNew YorkNYUSA
- The Third Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Yurii Chinenov
- Hospital for Special SurgeryNew YorkNYUSA
- David Z. Rosensweig Genomics Research CenterHospital for Special SurgeryNew YorkNYUSA
| | - Max Chao
- Hospital for Special SurgeryNew YorkNYUSA
- David Z. Rosensweig Genomics Research CenterHospital for Special SurgeryNew YorkNYUSA
| | | | | | - Matthew B Greenblatt
- Hospital for Special SurgeryNew YorkNYUSA
- Department of Pathology and Laboratory MedicineWeill Cornell MedicineNew YorkNYUSA
| | - Lionel B Ivashkiv
- Hospital for Special SurgeryNew YorkNYUSA
- David Z. Rosensweig Genomics Research CenterHospital for Special SurgeryNew YorkNYUSA
| | | | - Xu Yang
- Hospital for Special SurgeryNew YorkNYUSA
| |
Collapse
|
47
|
Andrés Sastre E, Nossin Y, Jansen I, Kops N, Intini C, Witte-Bouma J, van Rietbergen B, Hofmann S, Ridwan Y, Gleeson JP, O'Brien FJ, Wolvius EB, van Osch GJVM, Farrell E. A new semi-orthotopic bone defect model for cell and biomaterial testing in regenerative medicine. Biomaterials 2021; 279:121187. [PMID: 34678648 DOI: 10.1016/j.biomaterials.2021.121187] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/08/2021] [Accepted: 10/14/2021] [Indexed: 02/07/2023]
Abstract
In recent decades, an increasing number of tissue engineered bone grafts have been developed. However, expensive and laborious screenings in vivo are necessary to assess the safety and efficacy of their formulations. Rodents are the first choice for initial in vivo screens but their size limits the dimensions and number of the bone grafts that can be tested in orthotopic locations. Here, we report the development of a refined murine subcutaneous model for semi-orthotopic bone formation that allows the testing of up to four grafts per mouse one order of magnitude greater in volume than currently possible in mice. Crucially, these defects are also "critical size" and unable to heal within the timeframe of the study without intervention. The model is based on four bovine bone implants, ring-shaped, where the bone healing potential of distinct grafts can be evaluated in vivo. In this study we demonstrate that promotion and prevention of ossification can be assessed in our model. For this, we used a semi-automatic algorithm for longitudinal micro-CT image registration followed by histological analyses. Taken together, our data supports that this model is suitable as a platform for the real-time screening of bone formation, and provides the possibility to study bone resorption, osseointegration and vascularisation.
Collapse
Affiliation(s)
- E Andrés Sastre
- Department of Oral and Maxillofacial Surgery, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Y Nossin
- Department of Otorhinolaryngology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - I Jansen
- Department of Oral and Maxillofacial Surgery, Erasmus University Medical Center, Rotterdam, the Netherlands; Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - N Kops
- Department of Orthopaedics and Sports Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - C Intini
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - J Witte-Bouma
- Department of Oral and Maxillofacial Surgery, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - B van Rietbergen
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Department of Orthopaedic Surgery, Research School CAPHRI, Maastricht University Medical Center, Maastricht, the Netherlands
| | - S Hofmann
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Y Ridwan
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - J P Gleeson
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - F J O'Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; SFI Advanced Materials and Bioengineering Research (AMBER) Center, Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland; Trinity Center for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
| | - E B Wolvius
- Department of Oral and Maxillofacial Surgery, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - G J V M van Osch
- Department of Otorhinolaryngology, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Orthopaedics and Sports Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Delft, the Netherlands
| | - E Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
48
|
Fahy N, Palomares Cabeza V, Lolli A, Witte-Bouma J, Merino A, Ridwan Y, Wolvius EB, Hoogduijn MJ, Farrell E, Brama PAJ. Chondrogenically Primed Human Mesenchymal Stem Cells Persist and Undergo Early Stages of Endochondral Ossification in an Immunocompetent Xenogeneic Model. Front Immunol 2021; 12:715267. [PMID: 34659205 PMCID: PMC8515138 DOI: 10.3389/fimmu.2021.715267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/10/2021] [Indexed: 11/18/2022] Open
Abstract
Tissue engineering approaches using progenitor cells such as mesenchymal stromal cells (MSCs) represent a promising strategy to regenerate bone. Previous work has demonstrated the potential of chondrogenically primed human MSCs to recapitulate the process of endochondral ossification and form mature bone in vivo, using immunodeficient xenogeneic models. To further the translation of such MSC-based approaches, additional investigation is required to understand the impact of interactions between human MSC constructs and host immune cells upon the success of MSC-mediated bone formation. Although human MSCs are considered hypoimmunogenic, the potential of chondrogenically primed human MSCs to induce immunogenic responses in vivo, as well as the efficacy of MSC-mediated ectopic bone formation in the presence of fully competent immune system, requires further elucidation. Therefore, the aim of this study was to investigate the capacity of chondrogenically primed human MSC constructs to persist and undergo the process of endochondral ossification in an immune competent xenogeneic model. Chondrogenically differentiated human MSC pellets were subcutaneously implanted to wild-type BALB/c mice and retrieved at 2 and 12 weeks post-implantation. The percentages of CD4+ and CD8+ T cells, B cells, and classical/non-classical monocyte subsets were not altered in the peripheral blood of mice that received chondrogenic MSC constructs compared to sham-operated controls at 2 weeks post-surgery. However, MSC-implanted mice had significantly higher levels of serum total IgG compared to sham-operated mice at this timepoint. Flow cytometric analysis of retrieved MSC constructs identified the presence of T cells and macrophages at 2 and 12 weeks post-implantation, with low levels of immune cell infiltration to implanted MSC constructs detected by CD45 and CD3 immunohistochemical staining. Despite the presence of immune cells in the tissue, MSC constructs persisted in vivo and were not degraded/resorbed. Furthermore, constructs became mineralised, with longitudinal micro-computed tomography imaging revealing an increase in mineralised tissue volume from 4 weeks post-implantation until the experimental endpoint at 12 weeks. These findings indicate that chondrogenically differentiated human MSC pellets can persist and undergo early stages of endochondral ossification following subcutaneous implantation in an immunocompetent xenogeneic model. This scaffold-free model may be further extrapolated to provide mechanistic insight to osteoimmunological processes regulating bone regeneration and homeostasis.
Collapse
Affiliation(s)
- Niamh Fahy
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Orthopaedics and Sports Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - Virginia Palomares Cabeza
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Center, Rotterdam, Netherlands.,Transplantation Institute, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands.,School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Andrea Lolli
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Center, Rotterdam, Netherlands
| | - Janneke Witte-Bouma
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Center, Rotterdam, Netherlands
| | - Ana Merino
- Transplantation Institute, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - Yanto Ridwan
- Department of Genetics, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Eppo B Wolvius
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Center, Rotterdam, Netherlands
| | - Martin J Hoogduijn
- Transplantation Institute, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Center, Rotterdam, Netherlands
| | - Pieter A J Brama
- School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
49
|
Cai B, Lin D, Li Y, Wang L, Xie J, Dai T, Liu F, Tang M, Tian L, Yuan Y, Kong L, Shen SGF. N2-Polarized Neutrophils Guide Bone Mesenchymal Stem Cell Recruitment and Initiate Bone Regeneration: A Missing Piece of the Bone Regeneration Puzzle. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100584. [PMID: 34382372 PMCID: PMC8498914 DOI: 10.1002/advs.202100584] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/25/2021] [Indexed: 05/14/2023]
Abstract
The role of neutrophils in bone regeneration remains elusive. In this study, it is shown that intramuscular implantation of interleukin-8 (IL-8) (commonly recognized as a chemotactic cytokine for neutrophils) at different levels lead to outcomes resembling those of fracture hematoma at various stages. Ectopic endochondral ossification is induced by certain levels of IL-8, during which neutrophils are recruited to the implanted site and are N2-polarized, which then secrete stromal cell-derived factor-1α (SDF-1α) for bone mesenchymal stem cell (BMSC) chemotaxis via the SDF-1/CXCR4 (C-X-C motif chemokine receptor 4) axis and its downstream phosphatidylinositol 3'-kinase (PI3K)/Akt pathway and β-catenin-mediated migration. Neutrophils are pivotal for recruiting and orchestrating innate and adaptive immunocytes, as well as BMSCs at the initial stage of bone healing and regeneration. The results in this study delineate the mechanism of neutrophil-initiated bone regeneration and interaction between neutrophils and BMSCs, and innate and adaptive immunities. This work lays the foundation for research in the fields of bone regenerative therapy and biomaterial development, and might inspire further research into novel therapeutic options.
Collapse
Affiliation(s)
- Bolei Cai
- Department of Oral & Cranio‐Maxillofacial SurgeryShanghai Ninth People's HospitalCollege of StomatologyShanghai Jiao Tong University School of MedicineNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of Stomatology & Shanghai Research Institute of StomatologyShanghai200011China
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral DiseasesDepartment of Oral and Maxillofacial SurgerySchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Dan Lin
- Department of Oral & Cranio‐Maxillofacial SurgeryShanghai Ninth People's HospitalCollege of StomatologyShanghai Jiao Tong University School of MedicineNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of Stomatology & Shanghai Research Institute of StomatologyShanghai200011China
| | - Yan Li
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral DiseasesDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Le Wang
- Department of Oral & Cranio‐Maxillofacial SurgeryShanghai Ninth People's HospitalCollege of StomatologyShanghai Jiao Tong University School of MedicineNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of Stomatology & Shanghai Research Institute of StomatologyShanghai200011China
| | - Jirong Xie
- Department of Oral & Cranio‐Maxillofacial SurgeryShanghai Ninth People's HospitalCollege of StomatologyShanghai Jiao Tong University School of MedicineNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of Stomatology & Shanghai Research Institute of StomatologyShanghai200011China
- Department of ProsthodonticsSchool of Stomatologythe Jiamusi UniversityJiamusi154003China
| | - Taiqiang Dai
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral DiseasesDepartment of Oral and Maxillofacial SurgerySchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Fuwei Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral DiseasesDepartment of Oral and Maxillofacial SurgerySchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Mingyue Tang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral DiseasesDepartment of Oral and Maxillofacial SurgerySchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Lei Tian
- Department of Oral & Cranio‐Maxillofacial SurgeryShanghai Ninth People's HospitalCollege of StomatologyShanghai Jiao Tong University School of MedicineNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of Stomatology & Shanghai Research Institute of StomatologyShanghai200011China
| | - Yuan Yuan
- Key Laboratory for Ultrafine Materials of Ministry of EducationSchool of Materials Science and Engineeringand Engineering Research Center for Biomedical Materials of Ministry of EducationEast China University of Science and TechnologyShanghai200237P. R. China
| | - Liang Kong
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral DiseasesDepartment of Oral and Maxillofacial SurgerySchool of StomatologyThe Fourth Military Medical UniversityXi'an710032China
| | - Steve G. F. Shen
- Department of Oral & Cranio‐Maxillofacial SurgeryShanghai Ninth People's HospitalCollege of StomatologyShanghai Jiao Tong University School of MedicineNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of Stomatology & Shanghai Research Institute of StomatologyShanghai200011China
- Shanghai University of Medicine and Health SciencesShanghai201318P. R. China
| |
Collapse
|
50
|
Schlundt C, Fischer H, Bucher CH, Rendenbach C, Duda GN, Schmidt-Bleek K. The multifaceted roles of macrophages in bone regeneration: A story of polarization, activation and time. Acta Biomater 2021; 133:46-57. [PMID: 33974949 DOI: 10.1016/j.actbio.2021.04.052] [Citation(s) in RCA: 184] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/26/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022]
Abstract
To present knowledge, macrophages are found in all tissues of the human body. They are a cell population with high plasticity which come with a multitude of functions which appear to be adapted to the respective tissue niche and micro-environment in which they reside. Bone harbors multiple macrophage subpopulations, with the osteoclasts as classical representative of a bone resorbing cells and osteomacs as a bone tissue resident macrophage first described by the expression of F4/80. Both subtypes are found throughout all phases in bone healing. In vivo data on bone regeneration have demonstrated their essential role in initiating the healing cascade (inflammatory phase) but also of the later phases of healing (e.g. endochondral and intramembranous bone formation). To participate in such diverse processes macrophages have to be highly plastic in their functionality. Thus, the widely used M1/M2 paradigm to distinguish macrophage subpopulations may not mirror the comprehensive role of the dynamics of macrophage plasticity. From a clinical perspective it is especially relevant to distinguish what drives macrophages in impaired healing scenarios, implant loosening or infections, where their specific role of a misbalanced inflammatory setting is so far only partially known. With this review we aim at illustrating current knowledge and gaps of knowledge on macrophage plasticity and function during the cascades of regeneration and reconstitution of bone tissue. We propose aspects of the known biological mechanisms of macrophages and their specific subsets that might serve as targets to control their function in impaired healing and eventually support a scar-free regeneration. STATEMENT OF SIGNIFICANCE: Macrophages are essential for successful regeneration. In scar-free healing such as in bone, a complete failure of healing was shown if macrophages were depleted; the M1/M2 switch appears to be key to the progression from pro-inflammation to regeneration. However, experimental data illustrate that the classical M1/M2 paradigm does not completely mirror the complexity of observed macrophage functions during bone healing and thus demands a broader perspective. Within this review we discuss the high degree of plasticity of macrophages and the relevant contribution of the different and more specific M2 subtypes (M2a-M2f) during (bone) regeneration. It summarizes the versatile roles of macrophages in skeletal regeneration and thereby highlights potential target points for immunomodulatory approaches to enable or even foster bone repair.
Collapse
|