1
|
Wychowaniec JK, Bektas EI, Muerner M, Sapudom J, Šrejber M, Airoldi M, Schmidt R, Vernengo AJ, Edwards-Gayle CJC, Tipay PS, Otyepka M, Teo J, Eglin D, D'Este M. Effect of Tyrosine-Containing Self-Assembling β-Sheet Peptides on Macrophage Polarization and Inflammatory Response. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40235215 DOI: 10.1021/acsami.4c19900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Self-assembling peptides (SAPs) are fully defined nanobiomaterials offering unprecedented opportunities to control nanostructure and chemical attributes to investigate and manipulate cellular signals. To investigate the influence of chemical and morphological characteristics on inflammatory signaling in native immunity, we designed five β-sheet SAPs: EFEFKFEFK (EF8), YEFEFKFEFK (YEF8), EFEFKFEFKY (EF8Y), YEFEFKFEFKY (YEF8Y), and EYEFKFEFK (EYF8) (F: phenylalanine; E: glutamic acid; K: lysine, Y: tyrosine). The position of tyrosine in the peptide sequence dictated the self-assembly into nanostructures, with all SAPs self-assembling into thin constituent nanofibers with d ≈ 3.8 ± 0.4 nm, and sequences YEF8 and EF8 showing a propensity for associative bundling. These distinct SAPs induced contrasting inflammatory responses of monocytic model THP-1 cells-derived macrophages (MΦs). Presence of soluble EF8 nanofibers (at 2 mM) induced an anti-inflammatory response and polarization toward an M2 state, whereas YEF8 (at 2 mM) displayed a tendency for inducing a pro-inflammatory response and polarization toward an M1 state. EF8Y, YEF8Y, and EYF8 SAPs did not induce an inflammatory response in our models. These results were validated using peripheral blood mononuclear cells (PBMCs)-derived MΦs from human donors, confirming the critical role of EF8 and YEF8 SAPs as possible orchestrators of the repair of tissues or inducers of pro-inflammatory state, respectively. The same MΦs polarization responses from THP-1-derived MΦs cultured on 20 mM hydrogels were obtained. These findings will facilitate the utilization of this family of SAPs as immunomodulatory nanobiomaterials potentially changing the course of inflammation during the progression of various diseases.
Collapse
Affiliation(s)
| | - Ezgi Irem Bektas
- AO Research Institute Davos, Clavadelerstrasse 8, Davos 7270, Switzerland
| | - Marcia Muerner
- AO Research Institute Davos, Clavadelerstrasse 8, Davos 7270, Switzerland
- ETH Zürich, Rämistrasse 101, Zürich 8092, Switzerland
| | - Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - Martin Šrejber
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN), Palacký University Olomouc, 779 00 Olomouc, Czech Republic
| | - Marielle Airoldi
- AO Research Institute Davos, Clavadelerstrasse 8, Davos 7270, Switzerland
| | - Roland Schmidt
- Hitachi High-Tech Europe GmbH, Europark Fichtenhain A12, 47807 Krefeld, Germany
| | - Andrea J Vernengo
- AO Research Institute Davos, Clavadelerstrasse 8, Davos 7270, Switzerland
| | | | - Paul Sean Tipay
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - Michal Otyepka
- Regional Centre of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN), Palacký University Olomouc, 779 00 Olomouc, Czech Republic
- IT4Innovations, VSB-Technical University of Ostrava, 708 00 Ostrava-Poruba, Czech Republic
| | - Jeremy Teo
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - David Eglin
- Mines Saint-Étienne, Univ Jean Monnet, INSERM, UMR 1059 Sainbiose, 1059, Saint-Étienne, France
| | - Matteo D'Este
- AO Research Institute Davos, Clavadelerstrasse 8, Davos 7270, Switzerland
| |
Collapse
|
2
|
Riesner K, Hammerich L, Jahn D, Ellinghaus A, Polenz D, Grohmann L, Unger JK. What must not be named : Facts, emotions and coping strategies to enhance animal welfare in research. EMBO Rep 2025; 26:1929-1934. [PMID: 40164901 PMCID: PMC12019234 DOI: 10.1038/s44319-025-00429-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/12/2025] [Indexed: 04/02/2025] Open
Abstract
Scientists working with animals must maintain a delicate balance between conducting high-quality science and legal and ethical responsibilities. It is time to openly discuss their workload and emotional burden to improve both animal welfare and scientific quality in biomedical research.
Collapse
Affiliation(s)
- Katarina Riesner
- Taskforce Refinement, Charité - Univeristätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universitätsmedizin Berlin, Berlin, Germany
- Department of Hematology, Oncology and Tumorimmunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Linda Hammerich
- Taskforce Refinement, Charité - Univeristätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universitätsmedizin Berlin, Berlin, Germany
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Denise Jahn
- Taskforce Refinement, Charité - Univeristätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universitätsmedizin Berlin, Berlin, Germany
- Department of Oral and Maxillofacial Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Julius Wolff Institute, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Agnes Ellinghaus
- Taskforce Refinement, Charité - Univeristätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universitätsmedizin Berlin, Berlin, Germany
- Julius Wolff Institute, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Dietrich Polenz
- Taskforce Refinement, Charité - Univeristätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universitätsmedizin Berlin, Berlin, Germany
- Department of Surgery, Experimental Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Lisa Grohmann
- Taskforce Refinement, Charité - Univeristätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universitätsmedizin Berlin, Berlin, Germany.
- Charité 3R, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| | - Juliane K Unger
- Taskforce Refinement, Charité - Univeristätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universitätsmedizin Berlin, Berlin, Germany.
- Research Facilities of Experimental Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
3
|
Wychowaniec JK, Bektas EI, Vernengo AJ, Muerner M, Airoldi M, Tipay PS, Sapudom J, Teo J, Eglin D, D'Este M. Effect of molecular weight of tyramine-modified hyaluronan on polarization state of THP-1 and peripheral blood mononuclear cells-derived macrophages. BIOMATERIALS ADVANCES 2025; 169:214166. [PMID: 39823943 DOI: 10.1016/j.bioadv.2024.214166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 10/31/2024] [Accepted: 12/27/2024] [Indexed: 01/20/2025]
Abstract
The immunomodulatory properties of hyaluronan and its derivatives are key to their use in medicine and tissue engineering. In this work we evaluated the capability of soluble tyramine-modified hyaluronan (THA) synthesized from hyaluronan of two molecular weights (low Mw = 280 kDa and high Mw = 1640 kDa) for polarization of THP-1 and peripheral blood mononuclear cells (PBMCs)-derived macrophages (MΦs). We demonstrate the polarization effects of the supplemented THA by flow cytometry and bead-based multiplex immunoassay for the THP-1 derived MΦs and by semi-automated image analysis from confocal microscopy, immunofluorescent staining utilizing CD68 and CD206 surface markers, RT-qPCR gene expression analysis, as well as using the enzyme-linked immunosorbent assay (ELISA) for PBMCs-derived MΦs. Our data indicate that supplementation with LMW THA drives changes in THP-1 derived MΦs towards a pro-inflammatory M1-like phenotype, whereas supplementation with the HMW THA leads to a more mixed profile with some features of both M1 and M2 phenotypes, suggesting either a heterogeneous population or a transitional state. For cells directly sourced from human patients, PMBCs-derived MΦs, results exhibit a higher degree of variability, pointing out a differential regulation of factors including IL-10 and CD206 between the two cell sources. While human primary cells add to the clinical relevance, donor diversity introduces wider variability in the dataset, preventing drawing strong conclusions. Nevertheless, the MΦs profiles observed in THP-1 derived cells for treatments with LMW and HMW THA are generally consistent with what might be expected for the treatment with non-modified hyaluronans of respective molecular weights, confirming the known association holds true for the chemically tyramine-modified hyaluronan. We stipulate that these responses will provide basis for more accurate in vivo representation and translational immunomodulatory guidance for the use of THA-based biomaterials to a wider biomaterials and tissue engineering communities.
Collapse
Affiliation(s)
| | - Ezgi Irem Bektas
- AO Research Institute Davos, Clavadelerstrasse 8, Davos 7270, Switzerland
| | - Andrea J Vernengo
- AO Research Institute Davos, Clavadelerstrasse 8, Davos 7270, Switzerland
| | - Marcia Muerner
- AO Research Institute Davos, Clavadelerstrasse 8, Davos 7270, Switzerland; ETH Zürich, Rämistrasse 101, Zürich 8092, Switzerland
| | - Marielle Airoldi
- AO Research Institute Davos, Clavadelerstrasse 8, Davos 7270, Switzerland
| | | | - Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - Jeremy Teo
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - David Eglin
- Mines Saint-Étienne, Univ Jean Monnet, INSERM, U1059 Sainbiose, Saint-Étienne, France
| | - Matteo D'Este
- AO Research Institute Davos, Clavadelerstrasse 8, Davos 7270, Switzerland
| |
Collapse
|
4
|
Tchouto MN, Bucher CH, Mess AK, Haas S, Schmidt-Bleek K, Duda GN, Beule D, Milek M. Pronounced impairment of B cell differentiation during bone regeneration in adult immune experienced mice. Front Immunol 2025; 16:1511902. [PMID: 40098964 PMCID: PMC11911212 DOI: 10.3389/fimmu.2025.1511902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction Alterations of the adaptive immune system have been shown to impact bone healing and may result in impaired healing in some patients. Apart from T cells, B cells are the key drivers of adaptive immunity. Therefore, their role in age-associated impairments of bone healing might be essential to understand delays during the healing process. B cells are essential for bone formation, and their dysfunction has been associated with aging or autoimmune diseases. But whether age-associated changes in B cell phenotypes are involved in bone regeneration is unknown. Methods Here, we aimed to characterize the role of immune aging in B cell phenotypes during the early inflammatory phase of bone healing. By comparing non-immune experienced with young and immune experienced mice we aimed to analyze the effect of gained immune experience on B cells. Our single cell proteo-genomics analysis quantified thousands of transcriptomes of cells that were isolated from post osteotomy hematoma and the proximal and distal bone marrow cavities, and enabled us to evaluate cell proportion, differential gene expression and cell trajectories. Results While the B cell proportion in young and non-immune experienced animals did not significantly change from 2 to 5 days post osteotomy in the hematoma, we found a significant decrease of the B cell proportion in the immune experienced mice, which was accompanied by the decreased expression of B cell specific genes, suggesting a specific response in immune experienced animals. Furthermore, we detected the most extensive B cell differentiation block in immune-experienced mice compared to non-immune experienced and young animals, predominantly in the transition from immature to mature B cells. Discussion Our results suggest that the pronounced impairment of B cell production found in immune experienced animals plays an important role in the initial phase leading to delayed bone healing. Therefore, novel therapeutic approaches may be able target the B cell differentiation defect to retain B cell functionality even in the immune experienced setting, which is prone to delayed healing.
Collapse
Affiliation(s)
- Mireille Ngokingha Tchouto
- Julius Wolff Institute of Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Core Unit Bioinformatics, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christian H Bucher
- Julius Wolff Institute of Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ann-Kathrin Mess
- Julius Wolff Institute of Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Simon Haas
- Systems Hematology, Stem Cells & Precision Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute of Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Georg N Duda
- Julius Wolff Institute of Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Miha Milek
- Core Unit Bioinformatics, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
5
|
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
|
6
|
Arlt E, Kindermann A, Fritsche AK, Navarrete Santos A, Kielstein H, Bazwinsky-Wutschke I. A Flow Cytometry-Based Examination of the Mouse White Blood Cell Differential in the Context of Age and Sex. Cells 2024; 13:1583. [PMID: 39329764 PMCID: PMC11430320 DOI: 10.3390/cells13181583] [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: 07/31/2024] [Revised: 09/10/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
Analysis of the white blood cell differential as part of a flow cytometry-based approach is a common routine diagnostic tool used in clinics and research. For human blood, the methodological approach, suitable markers, and gating strategies are well-established. However, there is a lack of information regarding the mouse blood count. In this article, we deliver a fast and easy protocol for reprocessing mouse blood for the purpose of flow cytometric analysis, as well as suitable markers and gating strategies. We also present two possible applications: for the analysis of the whole blood count, with blood from a cardiac puncture, and for the analysis of a certain leukocyte subset at multiple time points in the framework of a mouse experiment, using blood from the facial vein. Additionally, we provide orientation values by applying the method to 3-month-old and 24-month-old male and female C57BL/6J mice. Our analyses demonstrate differences in the leukocyte fractions depending on age and sex. We discuss the influencing factors and limitations that can affect the results and that, therefore, need to be considered when applying this method. The present study fills the gap in the knowledge related to the rare information on flow cytometric analysis of mouse blood and, thus, lays the foundation for further investigations in this area.
Collapse
Affiliation(s)
- Elise Arlt
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin-Luther-University Halle-Wittenberg, 06108 Halle (Saale), Germany; (A.K.); (A.-K.F.); (H.K.); (I.B.-W.)
| | - Andrea Kindermann
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin-Luther-University Halle-Wittenberg, 06108 Halle (Saale), Germany; (A.K.); (A.-K.F.); (H.K.); (I.B.-W.)
| | - Anne-Kristin Fritsche
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin-Luther-University Halle-Wittenberg, 06108 Halle (Saale), Germany; (A.K.); (A.-K.F.); (H.K.); (I.B.-W.)
- Institute of Anatomy, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany
| | - Alexander Navarrete Santos
- Core Facility Flow Cytometry, Center for Basic Medical Research, Medical Faculty, Martin-Luther-University Halle-Wittenberg, 06108 Halle (Saale), Germany;
| | - Heike Kielstein
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin-Luther-University Halle-Wittenberg, 06108 Halle (Saale), Germany; (A.K.); (A.-K.F.); (H.K.); (I.B.-W.)
| | - Ivonne Bazwinsky-Wutschke
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin-Luther-University Halle-Wittenberg, 06108 Halle (Saale), Germany; (A.K.); (A.-K.F.); (H.K.); (I.B.-W.)
| |
Collapse
|
7
|
Yang W, Wang M, Hu J, Mo K, Xie X. The complex association between the immune system and the skeletal system in osteoporosis: A study of single-cell RNA sequencing. Int Immunopharmacol 2024; 138:112611. [PMID: 38976947 DOI: 10.1016/j.intimp.2024.112611] [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: 03/27/2024] [Revised: 06/28/2024] [Accepted: 06/30/2024] [Indexed: 07/10/2024]
Abstract
OBJECTIVE Osteoporosis (OP) is a disease characterized by decreased bone mass, deteriorated microstructure, and increased fragility and fracture risk. The diagnosis and prevention of OP and its complications have become major public health challenges. Therefore, exploring the complex ecological connections between the immune and skeletal systems may provide new insights for clinical prevention and treatment strategies. METHODS First, we performed single-cell RNA sequencing on human lumbar lamina tissue and conducted clustering and subgroup analysis of quality-controlled single-cell transcriptome data to identify target subgroups. Subsequently, enrichment analysis and pseudotime analysis were performed. In addition, we conducted in-depth studies on the gene regulatory network between different cell subgroups and the communication between bone immune cells. RESULTS In this study, we identified several cell subgroups that may be involved in the progression of OP. For example, the CCL4+ NKT and CXCL8+ neutrophils subgroups promote OP progression by mediating an inflammatory environment that disrupts bone homeostasis, and the MNDA+ Mac subgroup promotes osteoclast differentiation to promote OP. Moreover, the TNFAIP6+ Obl, NR4A2+ B and HMGN2+ erythrocyte subgroups promoted the balance of bone metabolism and suppressed OP. In the cell communication network, Obl closely interacts with immune cell subgroups through the CXCR4-CXCL12, CTGF-ITGB2, and TNFSF14-TNFRSF14 axes. CONCLUSION Our research revealed specific subgroups and intercellular interactions that play crucial roles in the pathogenesis of OP, providing potential new insights for more precise therapeutic interventions for OP.
Collapse
Affiliation(s)
- Weiwei Yang
- Department of Gynecology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi 545005, People's Republic of China
| | - Mingbo Wang
- Department of Orthopedics, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi 545005, People's Republic of China
| | - Juzheng Hu
- Department of Orthopedics, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi 545005, People's Republic of China; Department of Orthopedics, Liuzhou Worker's Hospital, Liuzhou, Guangxi 545005, People's Republic of China
| | - Ke Mo
- Clinical Research Center, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, People's Republic of China; Systems Biology Research Center, Biology Institute, Guangxi Academy of Sciences, Nanning 530007, Guangxi, People's Republic of China
| | - Xiangtao Xie
- Department of Orthopedics, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi 545005, People's Republic of China; Department of Orthopedics, Liuzhou Worker's Hospital, Liuzhou, Guangxi 545005, People's Republic of China.
| |
Collapse
|
8
|
Flanagan SD, Hougland JR, Zeng X, Cantrell PS, Sun M, Jones-Laughner J, Canino MC, Hughes JM, Foulis SA, Taylor KM, Walker LA, Guerriere KI, Sterczala AJ, Connaboy C, Beckner ME, Matheny RW, Nindl BC. Urinary Proteomic Biomarkers of Trabecular Bone Volume Change during Army Basic Combat Training. Med Sci Sports Exerc 2024; 56:1644-1654. [PMID: 38758530 DOI: 10.1249/mss.0000000000003464] [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: 05/18/2024]
Abstract
PURPOSE The purpose of this study is to optimize a dMS-based urinary proteomic technique and evaluate the relationship between urinary proteome content and adaptive changes in bone microarchitecture during BCT. METHODS Urinary proteomes were analyzed with an optimized dMS technique in two groups of 13 recruits ( N = 26) at the beginning (Pre) and end (Post) of BCT. Matched by age (21 ± 4 yr), sex (16 W), and baseline tibial trabecular bone volume fractions (Tb.BV/TV), these groups were distinguished by the most substantial (High) and minimal (Low) improvements in Tb.BV/TV. Differential protein expression was analyzed with mixed permutation ANOVA and false discovery proportion-based adjustment for multiple comparisons. RESULTS Tibial Tb.BV/TV increased from pre- to post-BCT in High (3.30 ± 1.64%, P < 0.0001) but not Low (-0.35 ± 1.25%, P = 0.4707). The optimized dMS technique identified 10,431 peptides from 1368 protein groups that represented 165 integrative biological processes. Seventy-four urinary proteins changed from pre- to post-BCT ( P = 0.0019), and neutrophil-mediated immunity was the most prominent ontology. Two proteins (immunoglobulin heavy constant gamma 4 and C-type lectin domain family 4 member G) differed from pre- to post-BCT in High and Low ( P = 0.0006). CONCLUSIONS The dMS technique can identify more than 1000 urinary proteins. At least 74 proteins are responsive to BCT, and other principally immune system-related proteins show differential expression patterns that coincide with adaptive bone formation.
Collapse
Affiliation(s)
| | | | - Xuemei Zeng
- Biomedical Mass Spectrometry Center, University of Pittsburgh, Pittsburgh, PA
| | - Pamela S Cantrell
- Biomedical Mass Spectrometry Center, University of Pittsburgh, Pittsburgh, PA
| | - Mai Sun
- Biomedical Mass Spectrometry Center, University of Pittsburgh, Pittsburgh, PA
| | | | - Maria C Canino
- Department of Sports Medicine and Nutrition, School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA
| | - Julie M Hughes
- Military Performance Division, United States Army Research Institute of Environmental Medicine, Natick, MA
| | - Stephen A Foulis
- Military Performance Division, United States Army Research Institute of Environmental Medicine, Natick, MA
| | - Kathryn M Taylor
- Military Performance Division, United States Army Research Institute of Environmental Medicine, Natick, MA
| | - Leila A Walker
- Military Performance Division, United States Army Research Institute of Environmental Medicine, Natick, MA
| | - Katelyn I Guerriere
- Military Performance Division, United States Army Research Institute of Environmental Medicine, Natick, MA
| | - Adam J Sterczala
- Department of Sports Medicine and Nutrition, School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA
| | | | - Meaghan E Beckner
- Military Performance Division, United States Army Research Institute of Environmental Medicine, Natick, MA
| | - Ronald W Matheny
- Military Operational Medicine Research Program, Fort Detrick, MD
| | - Bradley C Nindl
- Department of Sports Medicine and Nutrition, School of Health and Rehabilitation Sciences, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
9
|
Tao H, Zhu P, Xia W, Chu M, Chen K, Wang Q, Gu Y, Lu X, Bai J, Geng D. The Emerging Role of the Mitochondrial Respiratory Chain in Skeletal Aging. Aging Dis 2024; 15:1784-1812. [PMID: 37815897 PMCID: PMC11272194 DOI: 10.14336/ad.2023.0924] [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: 08/03/2023] [Accepted: 09/24/2023] [Indexed: 10/12/2023] Open
Abstract
Maintenance of mitochondrial homeostasis is crucial for ensuring healthy mitochondria and normal cellular function. This process is primarily responsible for regulating processes that include mitochondrial OXPHOS, which generates ATP, as well as mitochondrial oxidative stress, apoptosis, calcium homeostasis, and mitophagy. Bone mesenchymal stem cells express factors that aid in bone formation and vascular growth. Positive regulation of hematopoietic stem cells in the bone marrow affects the differentiation of osteoclasts. Furthermore, the metabolic regulation of cells that play fundamental roles in various regions of the bone, as well as interactions within the bone microenvironment, actively participates in regulating bone integrity and aging. The maintenance of cellular homeostasis is dependent on the regulation of intracellular organelles, thus understanding the impact of mitochondrial functional changes on overall bone metabolism is crucially important. Recent studies have revealed that mitochondrial homeostasis can lead to morphological and functional abnormalities in senescent cells, particularly in the context of bone diseases. Mitochondrial dysfunction in skeletal diseases results in abnormal metabolism of bone-associated cells and a secondary dysregulated microenvironment within bone tissue. This imbalance in the oxidative system and immune disruption in the bone microenvironment ultimately leads to bone dysplasia. In this review, we examine the latest developments in mitochondrial respiratory chain regulation and its impacts on maintenance of bone health. Specifically, we explored whether enhancing mitochondrial function can reduce the occurrence of bone cell deterioration and improve bone metabolism. These findings offer prospects for developing bone remodeling biology strategies to treat age-related degenerative diseases.
Collapse
Affiliation(s)
- Huaqiang Tao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
| | - Pengfei Zhu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
| | - Wenyu Xia
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
| | - Miao Chu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
| | - Kai Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
| | - Qiufei Wang
- Department of Orthopedics, Changshu Hospital Affiliated to Soochow University, First People’s Hospital of Changshu City, Jiangsu, China.
| | - Ye Gu
- Department of Orthopedics, Changshu Hospital Affiliated to Soochow University, First People’s Hospital of Changshu City, Jiangsu, China.
| | - Xiaomin Lu
- Department of Oncology, Affiliated Haian Hospital of Nantong University, Jiangsu, China.
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui, China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Jiangsu, China.
| |
Collapse
|
10
|
Rinne C, Soultoukis GA, Oveisi M, Leer M, Schmidt-Bleek O, Burkhardt LM, Bucher CH, Moussa EA, Makhlouf M, Duda GN, Saraiva LR, Schmidt-Bleek K, Schulz TJ. Caloric restriction reduces trabecular bone loss during aging and improves bone marrow adipocyte endocrine function in male mice. Front Endocrinol (Lausanne) 2024; 15:1394263. [PMID: 38904042 PMCID: PMC11188307 DOI: 10.3389/fendo.2024.1394263] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/13/2024] [Indexed: 06/22/2024] Open
Abstract
Introduction Caloric restriction (CR) is a nutritional intervention that increases life expectancy while lowering the risk for cardio-metabolic disease. Its effects on bone health, however, remain controversial. For instance, CR has been linked to increased accumulation of bone marrow adipose tissue (BMAT) in long bones, a process thought to elicit detrimental effects on bone. Qualitative differences have been reported in BMAT in relation to its specific anatomical localization, subdividing it into physiological and potentially pathological BMAT. We here examine the local impact of CR on bone composition, microstructure and its endocrine profile in the context of aging. Methods Young and aged male C57Bl6J mice were subjected to CR for 8 weeks and were compared to age-matched littermates with free food access. We assessed bone microstructure and BMAT by micro-CT, bone fatty acid and transcriptomic profiles, and bone healing. Results CR increased tibial BMAT accumulation and adipogenic gene expression. CR also resulted in elevated fatty acid desaturation in the proximal and mid-shaft regions of the tibia, thus more closely resembling the biochemical lipid profile of the distally located, physiological BMAT. In aged mice, CR attenuated trabecular bone loss, suggesting that CR may revert some aspects of age-related bone dysfunction. Cortical bone, however, was decreased in young mice on CR and remained reduced in aged mice, irrespective of dietary intervention. No negative effects of CR on bone regeneration were evident in either young or aged mice. Discussion Our findings indicate that the timing of CR is critical and may exert detrimental effects on bone biology if administered during a phase of active skeletal growth. Conversely, CR exerts positive effects on trabecular bone structure in the context of aging, which occurs despite substantial accumulation of BMAT. These data suggest that the endocrine profile of BMAT, rather than its fatty acid composition, contributes to healthy bone maintenance in aged mice.
Collapse
Affiliation(s)
- Charlotte Rinne
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
| | - George A. Soultoukis
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München, Germany
| | - Masoome Oveisi
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München, Germany
| | - Marina Leer
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München, Germany
| | - Oskar Schmidt-Bleek
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Lisa-Marie Burkhardt
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, Berlin, Germany
| | - Christian H. Bucher
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | | | - 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
| | - Luis R. Saraiva
- Translation Medicine Division, Sidra Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - 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
| | - 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, Germany
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| |
Collapse
|
11
|
Mehl J, Farahani SK, Brauer E, Klaus‐Bergmann A, Thiele T, Ellinghaus A, Bartels‐Klein E, Koch K, Schmidt‐Bleek K, Petersen A, Gerhardt H, Vogel V, Duda GN. External Mechanical Stability Regulates Hematoma Vascularization in Bone Healing Rather than Endothelial YAP/TAZ Mechanotransduction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307050. [PMID: 38273642 PMCID: PMC10987120 DOI: 10.1002/advs.202307050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/21/2023] [Indexed: 01/27/2024]
Abstract
Bone fracture healing is regulated by mechanobiological cues. Both, extracellular matrix (ECM) deposition and microvascular assembly determine the dynamics of the regenerative processes. Mechanical instability as by inter-fragmentary shear or compression is known to influence early ECM formation and wound healing. However, it remains unclear how these external cues shape subsequent ECM and microvascular network assembly. As transcriptional coactivators, the mechanotransducers yes-associated protein 1 (YAP)/transcriptional coactivator with PDZ-binding motif (TAZ) translate physical cues into downstream signaling events, yet their role in sprouting angiogenesis into the hematoma after injury is unknown. Using bone healing as model system for scar-free regeneration, the role of endothelial YAP/TAZ in combination with tuning the extrinsic mechanical stability via fracture fixation is investigated. Extrinsically imposed shear across the gap delayed hematoma remodeling and shaped the morphology of early collagen fiber orientations and microvascular networks, suggesting that enhanced shear increased the nutrient exchange in the hematoma. In contrast, endothelial YAP/TAZ deletion has little impact on the overall vascularization of the fracture gap, yet slightly increases the collagen fiber deposition under semi-rigid fixation. Together, these data provide novel insights into the respective roles of endothelial YAP/TAZ and extrinsic mechanical cues in orchestrating the process of bone regeneration.
Collapse
Affiliation(s)
- Julia Mehl
- Julius Wolff InstituteBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
- Berlin Institute of Health Center for Regenerative TherapiesBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
- Laboratory of Applied MechanobiologyDepartment of Health Sciences and TechnologyETH ZurichZurich8092Switzerland
| | - Saeed Khomeijani Farahani
- Julius Wolff InstituteBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
- Berlin Institute of Health Center for Regenerative TherapiesBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
| | - Erik Brauer
- Julius Wolff InstituteBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
- Berlin Institute of Health Center for Regenerative TherapiesBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
| | - Alexandra Klaus‐Bergmann
- Integrative Vascular Biology LaboratoryMax‐Delbrück‐Center for Molecular Medicine (MDC) in the Helmholtz Association13125BerlinGermany
- German Center for Cardiovascular Research (DZHK)Partnersite Berlin10785BerlinGermany
| | - Tobias Thiele
- Julius Wolff InstituteBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
- Berlin Institute of Health Center for Regenerative TherapiesBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
| | - Agnes Ellinghaus
- Julius Wolff InstituteBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
- Berlin Institute of Health Center for Regenerative TherapiesBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
| | - Eireen Bartels‐Klein
- Integrative Vascular Biology LaboratoryMax‐Delbrück‐Center for Molecular Medicine (MDC) in the Helmholtz Association13125BerlinGermany
- German Center for Cardiovascular Research (DZHK)Partnersite Berlin10785BerlinGermany
| | - Katharina Koch
- Integrative Vascular Biology LaboratoryMax‐Delbrück‐Center for Molecular Medicine (MDC) in the Helmholtz Association13125BerlinGermany
- German Center for Cardiovascular Research (DZHK)Partnersite Berlin10785BerlinGermany
| | - Katharina Schmidt‐Bleek
- Julius Wolff InstituteBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
- Berlin Institute of Health Center for Regenerative TherapiesBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
| | - Ansgar Petersen
- Julius Wolff InstituteBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
- Berlin Institute of Health Center for Regenerative TherapiesBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
| | - Holger Gerhardt
- Integrative Vascular Biology LaboratoryMax‐Delbrück‐Center for Molecular Medicine (MDC) in the Helmholtz Association13125BerlinGermany
- German Center for Cardiovascular Research (DZHK)Partnersite Berlin10785BerlinGermany
| | - Viola Vogel
- Laboratory of Applied MechanobiologyDepartment of Health Sciences and TechnologyETH ZurichZurich8092Switzerland
| | - Georg N. Duda
- Julius Wolff InstituteBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
- Berlin Institute of Health Center for Regenerative TherapiesBerlin Institute of Health at Charité – Universitätsmedizin Berlin13353BerlinGermany
| |
Collapse
|
12
|
Azadani RN, Karbasi S, Poursamar A. Chitosan/MWCNTs nanocomposite coating on 3D printed scaffold of poly 3-hydroxybutyrate/magnetic mesoporous bioactive glass: A new approach for bone regeneration. Int J Biol Macromol 2024; 260:129407. [PMID: 38224805 DOI: 10.1016/j.ijbiomac.2024.129407] [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: 11/08/2023] [Revised: 12/20/2023] [Accepted: 01/09/2024] [Indexed: 01/17/2024]
Abstract
The utilization of 3D printing has become increasingly common in the construction of composite scaffolds. In this study, magnetic mesoporous bioactive glass (MMBG) was incorporated into polyhydroxybutyrate (PHB) to construct extrusion-based 3D printed scaffold. After fabrication of the PHB/MMBG composite scaffolds, they were coated with chitosan (Cs) and chitosan/multi-walled carbon nanotubes (Cs/MWCNTs) solutions utilizing deep coating method. FTIR was conducted to confirm the presence of Cs and MWCNTs on the scaffolds' surface. The findings of mechanical analysis illustrated that presence of Cs/MWCNTs on the composite scaffolds increases compressive young modulus significantly, from 16.5 to 42.2 MPa. According to hydrophilicity evaluation, not only MMBG led to decrease the contact angle of pure PHB but also scaffolds surface modification utilization of Cs and MWCNTs, the contact angle decreased significantly from 82.34° to 54.15°. Furthermore, investigation of cell viability, cell metabolism and inflammatory cytokines such as interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α) and interleukin-10 (IL-10) and transforming growth factor-beta (TGF-β) proved that the scaffolds not only do not stimulate the immune system, but also polarize macrophage cells from M1 phase to M2 phase. The present study highlights the suitability of 3D printed scaffold PHB/MMBG with Cs/MWCNTs coating for bone tissue engineering.
Collapse
Affiliation(s)
- Reyhaneh Nasr Azadani
- Department of Biomaterials and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Saeed Karbasi
- Department of Biomaterials and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Dental Implants Research Center, Dental Research Institute, School of Dentistry, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Ali Poursamar
- Department of Biomaterials and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
13
|
Tan J, Li S, Sun C, Bao G, Liu M, Jing Z, Fu H, Sun Y, Yang Q, Zheng Y, Wang X, Yang H. A Dose-Dependent Spatiotemporal Response of Angiogenesis Elicited by Zn Biodegradation during the Initial Stage of Bone Regeneration. Adv Healthc Mater 2024; 13:e2302305. [PMID: 37843190 DOI: 10.1002/adhm.202302305] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/09/2023] [Indexed: 10/17/2023]
Abstract
Zinc (Zn) plays a crucial role in bone metabolism and imbues biodegradable Zn-based materials with the ability to promote bone regeneration in bone trauma. However, the impact of Zn biodegradation on bone repair, particularly its influence on angiogenesis, remains unexplored. This study reveals that Zn biodegradation induces a consistent dose-dependent spatiotemporal response in angiogenesis,both in vivo and in vitro. In a critical bone defect model, an increase in Zn release intensity from day 3 to 10 post-surgery is observed. By day 10, the CD31-positive area around the Zn implant significantly surpasses that of the Ti implant, indicating enhanced angiogenesis. Furthermore,angiogenesis exhibits a distance-dependent pattern closely mirroring the distribution of Zn signals from the implant. In vitro experiments demonstrate that Zn extraction fosters the proliferation and migration of human umbilical vein endothelial cells and upregulates the key genes associated with tube formation, such as HIF-1α and VEGF-A, peaking at a concentration of 22.5 µM. Additionally, Zn concentrations within the range of 11.25-45 µM promote the polarization of M0-type macrophages toward the M2-type, while inhibiting polarization toward the M1-type. These findings provide essential insights into the biological effects of Zn on bone repair, shedding light on its potential applications.
Collapse
Affiliation(s)
- Junlong Tan
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, 37 Xueyuan Rd, Beijing, 100191, China
| | - Shuang Li
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, 37 Xueyuan Rd, Beijing, 100191, China
| | - Chaoyang Sun
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, 37 Xueyuan Rd, Beijing, 100191, China
| | - Guo Bao
- Department of Reproduction and Physiology, National Research Institute for Family Planning, Beijing, 100081, China
| | - Meijing Liu
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, 37 Xueyuan Rd, Beijing, 100191, China
| | - Zehao Jing
- Beijing Key Laboratory of Spinal Disease Research, Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, P. R. China
| | - Hanwei Fu
- School of Materials Science and Engineering, Beihang University, 37 Xueyuan Rd, Beijing, China
| | - Yanhua Sun
- Shandong Provincial Key Laboratory of Microparticles Drug Delivery Technology, Qilu Pharmaceutical Co. Ltd., Jinan, 250100, China
| | - Qingmin Yang
- Shandong Provincial Key Laboratory of Microparticles Drug Delivery Technology, Qilu Pharmaceutical Co. Ltd., Jinan, 250100, China
| | - Yufeng Zheng
- Beijing Advanced Innovation Center for Materials Genome Engineering and School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Xiaogang Wang
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, 37 Xueyuan Rd, Beijing, 100191, China
| | - Hongtao Yang
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, 37 Xueyuan Rd, Beijing, 100191, China
| |
Collapse
|
14
|
Qian G, Mao Y, Shuai Y, Zeng Z, Peng S, Shuai C. Enhancing bone scaffold interfacial reinforcement through in situ growth of metal-organic frameworks (MOFs) on strontium carbonate: Achieving high strength and osteoimmunomodulation. J Colloid Interface Sci 2024; 655:43-57. [PMID: 37925968 DOI: 10.1016/j.jcis.2023.10.133] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 10/19/2023] [Accepted: 10/25/2023] [Indexed: 11/07/2023]
Abstract
Bioceramics have been extensively used to improve osteogenesis of polymers because of their excellent bone-forming capabilities. However, the inadequate interfacial bonding between ceramics and polymers compromises their mechanical properties. In this study, zeolitic imidazolate framework-8 (ZIF-8) was grown in situ on strontium carbonate (SrCO3) to construct a core-shell SrCO3@ZIF-8, which was then added to poly-l-lactic acid (PLLA) to print a SrCO3@ZIF-8/PLLA composite scaffold using selective sintering technology. First, ZIF-8 characterized by its multiple organic ligands, forms a robust interface with PLLA. Second, SrCO3 characterized by its negative zeta potential in solution, exhibits the ability to adsorb positively charged zinc ions. This, in turn, promotes the in situ growth of ZIF-8 on SrCO3, eventually achieving perfect bonding between the second phase and the PLLA matrix. Our findings indicated that the composite scaffold exhibited the highest compressive strength (21.93 MPa) and significantly promoted the osteogenic differentiation of mouse mesenchymal stem cells. Moreover, the in vivo results established that the SrCO3@ZIF-8/PLLA scaffold significantly accelerated bone regeneration efficiency in rat femur defects. The prepared scaffold, with its favorable mechanical properties and osteogenic activity, shows considerable promise for applications in bone repair.
Collapse
Affiliation(s)
- Guowen Qian
- Institute of Additive Manufacturing, Jiangxi University of Science and Technology, Nanchang 330013, China.
| | - Yuqian Mao
- Institute of Additive Manufacturing, Jiangxi University of Science and Technology, Nanchang 330013, China
| | - Yang Shuai
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhikui Zeng
- Department of Orthopedics, The Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang 330006, China.
| | - Shuping Peng
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China; NHC Key Laboratory of Carcinogenesis of Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan 410013, China
| | - Cijun Shuai
- Institute of Additive Manufacturing, Jiangxi University of Science and Technology, Nanchang 330013, China; State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China.
| |
Collapse
|
15
|
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
|
16
|
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
|
17
|
Liu R, Wu S, Liu W, Wang L, Dong M, Niu W. microRNAs delivered by small extracellular vesicles in MSCs as an emerging tool for bone regeneration. Front Bioeng Biotechnol 2023; 11:1249860. [PMID: 37720323 PMCID: PMC10501734 DOI: 10.3389/fbioe.2023.1249860] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/21/2023] [Indexed: 09/19/2023] Open
Abstract
Bone regeneration is a dynamic process that involves angiogenesis and the balance of osteogenesis and osteoclastogenesis. In bone tissue engineering, the transplantation of mesenchymal stem cells (MSCs) is a promising approach to restore bone homeostasis. MSCs, particularly their small extracellular vesicles (sEVs), exert therapeutic effects due to their paracrine capability. Increasing evidence indicates that microRNAs (miRNAs) delivered by sEVs from MSCs (MSCs-sEVs) can alter gene expression in recipient cells and enhance bone regeneration. As an ideal delivery vehicle of miRNAs, MSCs-sEVs combine the high bioavailability and stability of sEVs with osteogenic ability of miRNAs, which can effectively overcome the challenge of low delivery efficiency in miRNA therapy. In this review, we focus on the recent advancements in the use of miRNAs delivered by MSCs-sEVs for bone regeneration and disorders. Additionally, we summarize the changes in miRNA expression in osteogenic-related MSCs-sEVs under different microenvironments.
Collapse
Affiliation(s)
| | | | | | | | - Ming Dong
- School of Stomatology, Dalian Medical University, Dalian, China
| | - Weidong Niu
- School of Stomatology, Dalian Medical University, Dalian, China
| |
Collapse
|
18
|
Zhang Z, Ji C, Wang D, Wang M, She X, Song D, Xu X, Zhang D. Maresin1: A multifunctional regulator in inflammatory bone diseases. Int Immunopharmacol 2023; 120:110308. [PMID: 37192551 DOI: 10.1016/j.intimp.2023.110308] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/26/2023] [Accepted: 05/05/2023] [Indexed: 05/18/2023]
Abstract
Inflammation plays a crucial role in the physical response to danger signals, the elimination of toxic stimuli, and the restoration of homeostasis. However, dysregulated inflammatory responses lead to tissue damage, and chronic inflammation can disrupt osteogenic-osteoclastic homeostasis, ultimately leading to bone loss. Maresin1 (MaR1), a member of the specialized pro-resolving mediators (SPMs) family, has been found to possess significant anti-inflammatory, anti-allergic, pro-hemolytic, pro-healing, and pain-relieving properties. MaR1 is synthesized by macrophages (Mφs) and omega-3 fatty acids, and it may have the potential to promote bone homeostasis and treat inflammatory bone diseases. MaR1 has been found to stimulate osteoblast proliferation through leucine-rich repeat G protein-coupled receptor 6 (LGR6). It also activates Mφ phagocytosis and M2-type polarization, which helps to control the immune system. MaR1 can regulate T cells to exert anti-inflammatory effects and inhibit neutrophil infiltration and recruitment. In addition, MaR1 is involved in antioxidant signaling, including nuclear factor erythroid 2-related factor 2 (NRF2). It has also been found to promote the autophagic behavior of periodontal ligament stem cells, stimulate Mφs against pathogenic bacteria, and regulate tissue regeneration and repair. In summary, this review provides new information and a comprehensive overview of the critical roles of MaR1 in inflammatory bone diseases, indicating its potential as a therapeutic approach for managing skeletal metabolism and inflammatory bone diseases.
Collapse
Affiliation(s)
- Zhanwei Zhang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China; Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Chonghao Ji
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China; Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | | | - Maoshan Wang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China; Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Xiao She
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China; Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Dawei Song
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xin Xu
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China; Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China.
| | - Dongjiao Zhang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, China; Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China; Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China.
| |
Collapse
|
19
|
Guerrero J, Maevskaia E, Ghayor C, Bhattacharya I, Weber FE. Influence of Scaffold Microarchitecture on Angiogenesis and Regulation of Cell Differentiation during the Early Phase of Bone Healing: A Transcriptomics and Histological Analysis. Int J Mol Sci 2023; 24:ijms24066000. [PMID: 36983073 PMCID: PMC10056849 DOI: 10.3390/ijms24066000] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
The early phase of bone healing is a complex and poorly understood process. With additive manufacturing, we can generate a specific and customizable library of bone substitutes to explore this phase. In this study, we produced tricalcium phosphate-based scaffolds with microarchitectures composed of filaments of 0.50 mm in diameter, named Fil050G, and 1.25 mm named Fil125G, respectively. The implants were removed after only 10 days in vivo followed by RNA sequencing (RNAseq) and histological analysis. RNAseq results revealed upregulation of adaptive immune response, regulation of cell adhesion, and cell migration-related genes in both of our two constructs. However, significant overexpression of genes linked to angiogenesis, regulation of cell differentiation, ossification, and bone development was observed solely in Fil050G scaffolds. Moreover, quantitative immunohistochemistry of structures positive for laminin revealed a significantly higher number of blood vessels in Fil050G samples. Furthermore, µCT detected a higher amount of mineralized tissue in Fil050G samples suggesting a superior osteoconductive potential. Hence, different filament diameters and distances in bone substitutes significantly influence angiogenesis and regulation of cell differentiation involved in the early phase of bone regeneration, which precedes osteoconductivity and bony bridging seen in later phases and as consequence, impacts the overall clinical outcome.
Collapse
Affiliation(s)
- Julien Guerrero
- Oral Biotechnology and Bioengineering, Center for Dental Medicine, University of Zurich, 8032 Zurich, Switzerland
| | - Ekaterina Maevskaia
- Oral Biotechnology and Bioengineering, Center for Dental Medicine, University of Zurich, 8032 Zurich, Switzerland
| | - Chafik Ghayor
- Oral Biotechnology and Bioengineering, Center for Dental Medicine, University of Zurich, 8032 Zurich, Switzerland
| | - Indranil Bhattacharya
- Oral Biotechnology and Bioengineering, Center for Dental Medicine, University of Zurich, 8032 Zurich, Switzerland
| | - Franz E Weber
- Oral Biotechnology and Bioengineering, Center for Dental Medicine, University of Zurich, 8032 Zurich, Switzerland
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
20
|
Hochmann S, Ou K, Poupardin R, Mittermeir M, Textor M, Ali S, Wolf M, Ellinghaus A, Jacobi D, Elmiger JAJ, Donsante S, Riminucci M, Schäfer R, Kornak U, Klein O, Schallmoser K, Schmidt-Bleek K, Duda GN, Polansky JK, Geissler S, Strunk D. The enhancer landscape predetermines the skeletal regeneration capacity of stromal cells. Sci Transl Med 2023; 15:eabm7477. [PMID: 36947595 DOI: 10.1126/scitranslmed.abm7477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Multipotent stromal cells are considered attractive sources for cell therapy and tissue engineering. Despite numerous experimental and clinical studies, broad application of stromal cell therapeutics is not yet emerging. A major challenge is the functional diversity of available cell sources. Here, we investigated the regenerative potential of clinically relevant human stromal cells from bone marrow (BMSCs), white adipose tissue, and umbilical cord compared with mature chondrocytes and skin fibroblasts in vitro and in vivo. Although all stromal cell types could express transcription factors related to endochondral ossification, only BMSCs formed cartilage discs in vitro that fully regenerated critical-size femoral defects after transplantation into mice. We identified cell type-specific epigenetic landscapes as the underlying molecular mechanism controlling transcriptional stromal differentiation networks. Binding sites of commonly expressed transcription factors in the enhancer and promoter regions of ossification-related genes, including Runt and bZIP families, were accessible only in BMSCs but not in extraskeletal stromal cells. This suggests an epigenetically predetermined differentiation potential depending on cell origin that allows common transcription factors to trigger distinct organ-specific transcriptional programs, facilitating forward selection of regeneration-competent cell sources. Last, we demonstrate that viable human BMSCs initiated defect healing through the secretion of osteopontin and contributed to transient mineralized bone hard callus formation after transplantation into immunodeficient mice, which was eventually replaced by murine recipient bone during final tissue remodeling.
Collapse
Affiliation(s)
- Sarah Hochmann
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| | - Kristy Ou
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), T Cell Epigenetics, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Rodolphe Poupardin
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| | - Michaela Mittermeir
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| | - Martin Textor
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Salaheddine Ali
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Institute for Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Martin Wolf
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| | - Agnes Ellinghaus
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Dorit Jacobi
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Juri A J Elmiger
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Samantha Donsante
- Department of Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy
| | - Mara Riminucci
- Department of Molecular Medicine, Sapienza University of Rome, 00189 Rome, Italy
| | - Richard Schäfer
- Institute for Transfusion Medicine and Immunohematology, Goethe University Hospital, German Red Cross Blood Service Baden-Württemberg-Hessen gGmbH, 60323 Frankfurt am Main, Germany
- Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg, 79106 Freiburg, Germany
| | - Uwe Kornak
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Institute for Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
- Institute of Human Genetics, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Oliver Klein
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
| | | | - Katharina Schmidt-Bleek
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Georg N Duda
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Augustenburger Platz 1, 13353 Berlin, Germany
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Julia K Polansky
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), T Cell Epigenetics, Augustenburger Platz 1, 13353 Berlin, Germany
- German Rheumatism Research Centre (DRFZ), 10117 Berlin, Germany
| | - Sven Geissler
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute (JWI), Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Center for Advanced Therapies (BECAT), Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Dirk Strunk
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| |
Collapse
|
21
|
Wolter A, Bucher CH, Kurmies S, Schreiner V, Konietschke F, Hohlbaum K, Klopfleisch R, Löhning M, Thöne-Reineke C, Buttgereit F, Huwyler J, Jirkof P, Rapp AE, Lang A. A buprenorphine depot formulation provides effective sustained post-surgical analgesia for 72 h in mouse femoral fracture models. Sci Rep 2023; 13:3824. [PMID: 36882427 PMCID: PMC9992384 DOI: 10.1038/s41598-023-30641-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 02/27/2023] [Indexed: 03/09/2023] Open
Abstract
Adequate pain management is essential for ethical and scientific reasons in animal experiments and should completely cover the period of expected pain without the need for frequent re-application. However, current depot formulations of Buprenorphine are only available in the USA and have limited duration of action. Recently, a new microparticulate Buprenorphine formulation (BUP-Depot) for sustained release has been developed as a potential future alternative to standard formulations available in Europe. Pharmacokinetics indicate a possible effectiveness for about 72 h. Here, we investigated whether the administration of the BUP-Depot ensures continuous and sufficient analgesia in two mouse fracture models (femoral osteotomy) and could, therefore, serve as a potent alternative to the application of Tramadol via the drinking water. Both protocols were examined for analgesic effectiveness, side effects on experimental readout, and effects on fracture healing outcomes in male and female C57BL/6N mice. The BUP-Depot provided effective analgesia for 72 h, comparable to the effectiveness of Tramadol in the drinking water. Fracture healing outcome was not different between analgesic regimes. The availability of a Buprenorphine depot formulation for rodents in Europe would be a beneficial addition for extended pain relief in mice, thereby increasing animal welfare.
Collapse
Affiliation(s)
- Angelique Wolter
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- German Rheumatism Research Centre (DRFZ), Leibniz Institute, Berlin, Germany.
- Institute of Animal Welfare, Animal Behavior and Laboratory Animal Science, Department of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany.
| | - Christian H Bucher
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Kurmies
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Centre (DRFZ), Leibniz Institute, Berlin, Germany
| | - Viktoria Schreiner
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Frank Konietschke
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Berlin, Germany
- Institute of Biometry and Clinical Epidemiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Katharina Hohlbaum
- Institute of Animal Welfare, Animal Behavior and Laboratory Animal Science, Department of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
- German Centre for the Protection of Laboratory Animals (Bf3R), German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - Robert Klopfleisch
- Institute of Veterinary Pathology, Department of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Max Löhning
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Centre (DRFZ), Leibniz Institute, Berlin, Germany
| | - Christa Thöne-Reineke
- Institute of Animal Welfare, Animal Behavior and Laboratory Animal Science, Department of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Frank Buttgereit
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Centre (DRFZ), Leibniz Institute, Berlin, Germany
| | - Jörg Huwyler
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Paulin Jirkof
- Office for Animal Welfare and 3Rs, University of Zurich, Zurich, Switzerland
| | - Anna E Rapp
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Centre (DRFZ), Leibniz Institute, Berlin, Germany
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Department of Orthopedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Annemarie Lang
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- German Rheumatism Research Centre (DRFZ), Leibniz Institute, Berlin, Germany.
- Departments of Orthopaedic Surgery and Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
22
|
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
|
23
|
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: 104] [Impact Index Per Article: 52.0] [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
|
24
|
Zhang X, Lin X, Wang M, Deng L, Wei L, Liu Y. Icariin Has a Synergistic Effect on the Osteoinductivity of Bone Morphogenetic Protein 2 at Ectopic Sites. Orthop Surg 2023; 15:540-548. [PMID: 36628510 PMCID: PMC9891965 DOI: 10.1111/os.13597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 10/22/2022] [Accepted: 08/07/2022] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVE Establishing biocompatible, biodegradable, osteoconductive, and osteoinductive bone materials remains a challenging subject in the research of bone healing and bone regeneration. Previously, we demonstrated the osteogenic and osteoconductive effects of biomimetic calcium phosphate (BioCaP) incorporating with Icariin and/or bone morphogenetic protein 2 (BMP-2) at orthotopic sites. METHODS By implanting the BioCaP granules incorporated Icariin and/or BMP-2 into the dorsal subcutaneous pockets of adult male Sprague-Dawley (S-D) rats (6-7 weeks old), we investigated the osteoinductive efficacy of the samples. Micro-computed tomography(micro-CT) observations and histological slices were used to verify the osteoinduction of this system on the 2nd and 5th week. Statistical significances was evaluated using Turkey's post hoc test of one-way analysis of variance. RESULTS The osteoinduction of the BioCaP incorporated with BMP-2 or both agents was confirmed as expected. BioCaP with Icariin alone could not generate bone formation at an ectopic sites. Nevertheless, co-administration of Icariin increased bone mineral density (BMD; p < 0.01) (628mg HA/cm3 vs 570mg HA/cm3 ) and completely changed the distribution of newly formed bone when compared with the granules with BMP-2 alone, even though there was no significant difference in the volume of newly formed bone. In contrast, the BioCaP with both agents (37.86%) had significantly fewer remaining materials than the other groups by the end of the fifth week (53.22%, 53.62% and 48.22%) (p < 0.01). CONCLUSION The co-administration of Icariin and BMP-2 increased BMD changed the distribution of newly formed bone, and reduced the amount of remaining materials. Therefore, Icariin can stimulate BMP-2 when incorporated into BioCaP granules at ectopic sites, which makes it useful for bone tissue engineering.
Collapse
Affiliation(s)
- Xin Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceCancer Center of Zhejiang UniversityHangzhouChina
| | - Xingnan Lin
- School of DentistryZhejiang Chinese Medical UniversityHangzhouChina
| | - Mingjie Wang
- Department of Oral Cell Biology, Academic Center of Dentistry (ACTA)University of Amsterdam and VU UniversityAmsterdamThe Netherlands
| | - Liquan Deng
- School of StomatologyZhejiang Chinese Medical UniversityHangzhouChina
| | - Lingfei Wei
- Department of Oral Cell Biology, Academic Center of Dentistry (ACTA)University of Amsterdam and VU UniversityAmsterdamThe Netherlands,Department of Dental ImplantologyYantai Stomatological HospitalYantaiChina
| | - Yuelian Liu
- Department of Oral Cell Biology, Academic Center of Dentistry (ACTA)University of Amsterdam and VU UniversityAmsterdamThe Netherlands
| |
Collapse
|
25
|
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
|
26
|
Xiong Y, Mi BB, Lin Z, Hu YQ, Yu L, Zha KK, Panayi AC, Yu T, Chen L, Liu ZP, Patel A, Feng Q, Zhou SH, Liu GH. The role of the immune microenvironment in bone, cartilage, and soft tissue regeneration: from mechanism to therapeutic opportunity. Mil Med Res 2022; 9:65. [PMID: 36401295 PMCID: PMC9675067 DOI: 10.1186/s40779-022-00426-8] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 10/30/2022] [Indexed: 11/21/2022] Open
Abstract
Bone, cartilage, and soft tissue regeneration is a complex spatiotemporal process recruiting a variety of cell types, whose activity and interplay must be precisely mediated for effective healing post-injury. Although extensive strides have been made in the understanding of the immune microenvironment processes governing bone, cartilage, and soft tissue regeneration, effective clinical translation of these mechanisms remains a challenge. Regulation of the immune microenvironment is increasingly becoming a favorable target for bone, cartilage, and soft tissue regeneration; therefore, an in-depth understanding of the communication between immune cells and functional tissue cells would be valuable. Herein, we review the regulatory role of the immune microenvironment in the promotion and maintenance of stem cell states in the context of bone, cartilage, and soft tissue repair and regeneration. We discuss the roles of various immune cell subsets in bone, cartilage, and soft tissue repair and regeneration processes and introduce novel strategies, for example, biomaterial-targeting of immune cell activity, aimed at regulating healing. Understanding the mechanisms of the crosstalk between the immune microenvironment and regeneration pathways may shed light on new therapeutic opportunities for enhancing bone, cartilage, and soft tissue regeneration through regulation of the immune microenvironment.
Collapse
Affiliation(s)
- Yuan Xiong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Bo-Bin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 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, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Yi-Qiang Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Le Yu
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, USA
| | - Kang-Kang Zha
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.,Key Laboratory of Biorheological Science and Technology,Ministry of Education College of Bioengineering, Chongqing University, Shapingba, Chongqing, 400044, China
| | - Adriana C Panayi
- Department of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02152, USA
| | - Tao Yu
- Department of Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lang Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.,Department of Physics, Center for Hybrid Nanostructure (CHyN), University of Hamburg, Hamburg, 22761, Germany
| | - Zhen-Ping Liu
- Department of Physics, Center for Hybrid Nanostructure (CHyN), University of Hamburg, Hamburg, 22761, Germany.,Joint Laboratory of Optofluidic Technology and System,National Center for International Research on Green Optoelectronics, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou, 510006, China
| | - Anish Patel
- Skeletal Biology Laboratory, Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02120, USA
| | - Qian Feng
- Key Laboratory of Biorheological Science and Technology,Ministry of Education College of Bioengineering, Chongqing University, Shapingba, Chongqing, 400044, China.
| | - Shuan-Hu Zhou
- Skeletal Biology Laboratory, Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02120, USA. .,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA.
| | - Guo-Hui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| |
Collapse
|
27
|
Khass M, Rashid H, Burrows PD, Javed A, Schroeder HW. Loss of early B cell protein λ5 decreases bone mass and accelerates skeletal aging. Front Immunol 2022; 13:906649. [PMID: 36189270 PMCID: PMC9516392 DOI: 10.3389/fimmu.2022.906649] [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] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
The early B cell protein λ5 is an essential component of the surrogate light chain and the preB cell receptor (preBCR), which is critical for optimal B cell development. To investigate the effect of λ5 and/or B cells on bone acquisition over time, we developed a panel of JH -/- , λ5-/-, JH -/- λ5-/-, and wild-type (WT) BALB/c mice and then studied postnatal bone development and aging in these mice at one, six, twelve, and twenty-two months of age. The trabecular bone volume over total volume (BV/TV) in JH -/- mice was similar to WT mice at all ages. In contrast, at six months of age and thereafter, λ5-/- and JH -/- λ5-/- mice demonstrated a severe decrease in trabecular bone mass. Surprisingly, bone mass in six-month-old λ5-/- and JH -/- λ5-/- mice was similar to or even lower than in aged (twenty-two-months) WT mice, suggesting accelerated skeletal aging. The postnatal development and the acquisition of cortical bone mass in JH -/- λ5-/- mice were generally comparable to WT. However, JH -/- λ5-/- mice showed a significant decrease in cortical BV/TV at six- and twelve months of age. To examine the contribution of λ5 and B cells to postnatal bone synthesis, we separately transplanted whole bone marrow cells from JH -/- λ5-/- and WT mice into irradiated JH -/- λ5-/- and WT recipients. WT recipients of JH -/- λ5-/- marrow cells failed to show acquisition of trabecular bone mass, whereas transplanting WT marrow cells into JH -/- λ5-/- recipients led to the recovery of trabecular bone mass. Transfer of WT marrow cells into JH -/- λ5-/- mice promoted synthesis of new cortical and trabecular bone. Our findings indicate that λ5 plays a major role in preserving bone mass during postnatal development and skeletal aging which is distinct from its role in B cell development. The absence of both λ5 and B cells in JH -/- λ5-/- mice leads to delayed acquisition of cortical bone during postnatal development. Dissecting the mechanism(s) by which λ5 regulates bone homeostasis may provide new avenues for the treatment of age-related loss of bone mass and osteoporosis.
Collapse
Affiliation(s)
- Mohamed Khass
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Harunur Rashid
- Department of Oral and Maxillofacial Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Peter D. Burrows
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Amjad Javed
- Department of Oral and Maxillofacial Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Harry W. Schroeder
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
28
|
Fretwurst T, Tritschler I, Rothweiler R, Nahles S, Altmann B, Schilling O, Nelson K. Proteomic profiling of human bone from different anatomical sites - A pilot study. Proteomics Clin Appl 2022; 16:e2100049. [PMID: 35462455 DOI: 10.1002/prca.202100049] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 04/11/2022] [Accepted: 04/20/2022] [Indexed: 12/30/2022]
Abstract
PURPOSE The study aim is a comparative proteome-based analysis of different autologous bone entities (alveolar bone [AB], iliac cortical [IC] bone, and iliac spongiosa [IS]) used for alveolar onlay grafting. EXPERIMENTAL DESIGN Site-matched bone samples of AB, IC, and IS were harvested during alveolar onlay grafting. Proteins were extracted using a detergent-based (sodium dodecyl sulfate) strategy and trypsinized. Proteome analysis was performed using liquid chromatography-tandem mass spectrometry (LC-MS/MS). MaxQuant was used for peptide-to-spectrum matching, peak detection, and quantitation. Linear models for microarray analysis (LIMMA) were used to detect differentially abundant peptides and proteins. RESULTS A total of 1730 different proteins were identified across the 15 samples at a false discovery rate of 1%. Partial least-squares discriminant analysis approved segregation of AB, IC, and IS protein profiles. LIMMA statistics highlighted 66 proteins that were more abundant in AB then in IC (vs. 92 proteins were enriched in IC over AB). Gene Ontology enrichment analysis revealed a matrisomal versus an immune-related proteome fingerprint in AB versus IC. CONCLUSION AND CLINICAL RELEVANCE This pilot study demonstrates an ECM protein-related proteome fingerprint in AB and an immune-related proteome fingerprint in IS and IC.
Collapse
Affiliation(s)
- Tobias Fretwurst
- Department of Oral- and Craniomaxillofacial Surgery/Translational Implantology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | | | - René Rothweiler
- Department of Oral- and Craniomaxillofacial Surgery/Translational Implantology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Susanne Nahles
- Department of Oral and Maxillofacial Surgery, Berlin Institute of Health, Corporate Member of Freie Universität Berlin, Charité - Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Brigitte Altmann
- Department of Prosthetic Dentistry, Center for Dental Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,G.E.R.N Center for Tissue Replacement, Regeneration & Neogenesis, Department of Prosthetic Dentistry, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Oliver Schilling
- Institute of Surgical Pathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Katja Nelson
- Department of Oral- and Craniomaxillofacial Surgery/Translational Implantology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| |
Collapse
|
29
|
Nadine S, Correia CR, Mano JF. Engineering immunomodulatory hydrogels and cell-laden systems towards bone regeneration. BIOMATERIALS ADVANCES 2022; 140:213058. [PMID: 35933955 DOI: 10.1016/j.bioadv.2022.213058] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/27/2022] [Accepted: 07/31/2022] [Indexed: 06/15/2023]
Abstract
The well-known synergetic interplay between the skeletal and immune systems has changed the design of advanced bone tissue engineering strategies. The immune system is essential during the bone lifetime, with macrophages playing multiple roles in bone healing and biomaterial integration. If in the past, the most valuable aspect of implants was to avoid immune responses of the host, nowadays, it is well-established how important are the crosstalks between immune cells and bone-engineered niches for an efficient regenerative process to occur. For that, it is essential to recapitulate the multiphenotypic cellular environment of bone tissue when designing new approaches. Indeed, the lack of osteoimmunomodulatory knowledge may be the explanation for the poor translation of biomaterials into clinical practice. Thus, smarter hydrogels incorporating immunomodulatory bioactive factors, stem cells, and immune cells are being proposed to develop a new generation of bone tissue engineering strategies. This review highlights the power of immune cells to upgrade the development of innovative engineered strategies, mainly focusing on orthopaedic and dental applications.
Collapse
Affiliation(s)
- Sara Nadine
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| | - Clara R Correia
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - João F Mano
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| |
Collapse
|
30
|
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
|
31
|
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
|
32
|
Khokhani P, Belluomo R, Croes M, Gawlitta D, Kruyt MC, Weinans H. An in-vitro model to test the influence of immune cell secretome on MSC osteogenic differentiation. Tissue Eng Part C Methods 2022; 28:420-430. [PMID: 35770885 DOI: 10.1089/ten.tec.2022.0086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Immune cells and their soluble factors have an important role in the bone healing process. Modulation of the immune response, therefore, offers a potential strategy to enhance bone formation. To investigate the influence of the immune system on osteogenesis, we developed and applied an in-vitro model that incorporates both innate and adaptive immune cells. Human peripheral blood mononuclear cells (PBMCs) were isolated and cultured for 24 hours and subsequently stimulated with immune-modulatory agents; C-class CpG oligodeoxynucleotide (CpG ODN C), Polyinosinic acid-polycytidylic acid Poly(I:C), and lipopolysaccharide (LPS); all pathogen recognition receptor agonists, and that target Toll-like receptors TLR9, -3, and -4, respectively. The conditioned medium obtained from PBMCs after 24 hours was used to investigate its effects on the metabolic activity and osteogenic differentiation capacity of human bone marrow-derived mesenchymal stromal cells (MSCs). Conditioned media from unstimulated PBMCs did not affect the metabolic activity and osteogenic differentiation capacity of MSCs. The conditioned medium from CpG ODN C and LPS stimulated PBMCs increased alkaline phosphatase activity of MSCs by approximately 3-fold as compared to the unstimulated control, whereas Poly(I:C) conditioned medium did not enhance ALP activity of MSCs. Moreover, direct stimulation of MSCs with the immune-modulatory stimuli did not result in increased alkaline phosphatase activity. These results demonstrate that soluble factors present in conditioned medium from PBMCs stimulated with immune-modulatory factors enhance osteogenesis of MSCs. This in-vitro model can serve as a tool in screening immune-modulatory stimulants from a broad variety of immune cells for (indirect) effects on osteogenesis and also to identify soluble factors from multiple immune cell types that may modulate bone healing.
Collapse
Affiliation(s)
- Paree Khokhani
- University Medical Centre Utrecht, 8124, Orthopedics , UMC Utrecht, dept. Orthopedics, G5.203, Heidelberglaan 100, Utrecht, Utrecht, Drenthe, Netherlands, 3584CX.,University Medical Centre, Utrecht (UMCU), UMC Utrecht, dept. Orthopedics, G5.203, Heidelberglaan 100, Netherlands;
| | - Ruggero Belluomo
- University Medical Centre Utrecht, 8124, Orthopedics , Utrecht, Utrecht, Netherlands;
| | - Michiel Croes
- University Medical Centre Utrecht, 8124, Orthopedics , Utrecht, Utrecht, Netherlands;
| | - Debby Gawlitta
- University Medical Center Utrecht, Oral and Maxillofacial Surgery, Prosthodontics & Special Dental Care, Heidelberglaan 100, G05.129, PO Box 85500, Utrecht, Netherlands, 3508 GA;
| | - Moyo C Kruyt
- University medical center Utrecht, Orthopedics, HP G 05.228, PO Box 85500, Utrecht, Netherlands, 3508 GA;
| | - Harrie Weinans
- University Medical Centre Utrecht, 8124, Orthopedics, Utrecht, Utrecht, Netherlands;
| |
Collapse
|
33
|
Jiang Q, Huang X, Yu W, Huang R, Zhao X, Chen C. mTOR Signaling in the Regulation of CD4+ T Cell Subsets in Periodontal Diseases. Front Immunol 2022; 13:827461. [PMID: 35222410 PMCID: PMC8866697 DOI: 10.3389/fimmu.2022.827461] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/20/2022] [Indexed: 02/05/2023] Open
Abstract
Periodontal disease results from the inflammatory infiltration by the microbial community which is marked through tooth mobility and alveolar bone resorption. The inflammation in periodontal disease is mediated by CD4+ T cells through cytokine secretion and osteoclastogenetic activity. Historically, the inflammatory model in periodontal disease is described through disruption of the balance between two subsets of T helper cells which are T-helper type 1 (Th1) and T-helper type 2 (Th2). However, more and more studies have found that apart from subsets of helper T cells, regulatory T-cells and Th17 cells are also involved in the pathogenesis of periodontal diseases. Growing evidence proves that helper T cells differentiation, activation, and subset determination are under the strong impact of mTOR signaling. mTOR signaling could promote Th1 and Th17 cell differentiation and inhibit Treg commitment through different mTOR complexes, therefore we anticipate a regulation effect of mTOR signaling on periodontal diseases by regulating CD4+ T cell subsets. This review aims to integrate the topical researches about the role of different types of Th cells in the pathogenesis of periodontal diseases, as well as the regulation of mTOR signaling in the specification and selection of Th cell commitment.
Collapse
Affiliation(s)
- Qian Jiang
- Department of Oral and Maxillofacial Surgery and Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Xiaobin Huang
- Department of Oral and Maxillofacial Surgery and Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Wenjing Yu
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ranran Huang
- Department of Oral and Maxillofacial Surgery and Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Xuefeng Zhao
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States.,State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chider Chen
- Department of Oral and Maxillofacial Surgery and Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Center of Innovation and Precision Dentistry, School of Dental Medicine, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
34
|
Dirnagl U, Duda GN, Grainger DW, Reinke P, Roubenoff R. Reproducibility, relevance and reliability as barriers to efficient and credible biomedical technology translation. Adv Drug Deliv Rev 2022; 182:114118. [PMID: 35066104 DOI: 10.1016/j.addr.2022.114118] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/14/2022] [Accepted: 01/15/2022] [Indexed: 12/23/2022]
Abstract
Biomedical research accuracy and relevance for improving healthcare are increasingly identified as costly problems. Basic research data quality, reporting and methodology, and reproducibility are common factors implicated in this challenge. Preclinical models of disease and therapy, largely conducted in rodents, have known deficiencies in replicating most human conditions. Their translation to human results is acknowledged to be poor for decades. Clinical data quality and quantity is also recognized as deficient; gold standard randomized clinical trials are expensive. Few solid conclusions from clinical studies are replicable and many remain unpublished. The translational pathway from fundamental biomedical research through to innovative solutions handed to clinical practitioners is therefore highly inefficient and costly in terms of wasted resources, early claims from fundamental discoveries never witnessed in humans, and few new, improved solutions available clinically for myriad diseases. Improving this biomedical research strategy and resourcing for reliability, translational relevance, reproducibility and clinical impact requires careful analysis and consistent enforcement at both funding and peer review levels.
Collapse
Affiliation(s)
- Ulrich Dirnagl
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Germany; QUEST Center for Responsible Research, Berlin Institute of Health, Germany
| | - Georg N Duda
- Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany; Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany
| | - David W Grainger
- Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, Salt Lake City, UT 84112 USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112 USA.
| | - Petra Reinke
- Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany; Berlin Center for Advanced Therapies (BeCAT), Charité - Universitaetsmedizin Berlin, 13353 Berlin, Germany
| | - Ronenn Roubenoff
- Novartis Institutes for Biomedical Research, Cambridge, Basel, Massachusetts, Switzerland
| |
Collapse
|
35
|
Lang A, Stefanowski J, Pfeiffenberger M, Wolter A, Damerau A, Hemmati-Sadeghi S, Haag R, Hauser AE, Löhning M, Duda GN, Hoff P, Schmidt-Bleek K, Gaber T, Buttgereit F. MIF does only marginally enhance the pro-regenerative capacities of DFO in a mouse-osteotomy-model of compromised bone healing conditions. Bone 2022; 154:116247. [PMID: 34743042 DOI: 10.1016/j.bone.2021.116247] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/23/2021] [Accepted: 10/25/2021] [Indexed: 12/18/2022]
Abstract
The initial phase of fracture healing is crucial for the success of bone regeneration and is characterized by an inflammatory milieu and low oxygen tension (hypoxia). Negative interference with or prolongation of this fine-tuned initiation phase will ultimately lead to a delayed or incomplete healing such as non-unions which then requires an effective and gentle therapeutic intervention. Common reasons include a dysregulated immune response, immunosuppression or a failure in cellular adaptation to the inflammatory hypoxic milieu of the fracture gap and a reduction in vascularizing capacity by environmental noxious agents (e.g. rheumatoid arthritis or smoking). The hypoxia-inducible factor (HIF)-1α is responsible for the cellular adaptation to hypoxia, activating angiogenesis and supporting cell attraction and migration to the fracture gap. Here, we hypothesized that stabilizing HIF-1α could be a cost-effective and low-risk prevention strategy for fracture healing disorders. Therefore, we combined a well-known HIF-stabilizer - deferoxamine (DFO) - and a less known HIF-enhancer - macrophage migration inhibitory factor (MIF) - to synergistically induce improved fracture healing. Stabilization of HIF-1α enhanced calcification and osteogenic differentiation of MSCs in vitro. In vivo, only the application of DFO without MIF during the initial healing phase increased callus mineralization and vessel formation in a preclinical mouse-osteotomy-model modified to display a compromised healing. Although we did not find a synergistically effect of MIF when added to DFO, our findings provide additional support for a preventive strategy towards bone healing disorders in patients with a higher risk by accelerating fracture healing using DFO to stabilize HIF-1α.
Collapse
Affiliation(s)
- Annemarie Lang
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany; Berlin Institute of Health at Charité Universitätsmedizin Berlin, Center for Regenerative Therapies, Berlin, Germany
| | - Jonathan Stefanowski
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Moritz Pfeiffenberger
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Angelique Wolter
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Alexandra Damerau
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Shabnam Hemmati-Sadeghi
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; Berlin Institute of Health at Charité Universitätsmedizin Berlin, Center for Regenerative Therapies, Berlin, Germany; Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Anja E Hauser
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Max Löhning
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Georg N Duda
- Berlin Institute of Health at Charité Universitätsmedizin Berlin, Center for Regenerative Therapies, Berlin, Germany; Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany; Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Julius Wolff Institute, Berlin, Germany
| | - Paula Hoff
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany; Berlin Institute of Health at Charité Universitätsmedizin Berlin, Center for Regenerative Therapies, Berlin, Germany
| | - Katharina Schmidt-Bleek
- Berlin Institute of Health at Charité Universitätsmedizin Berlin, Center for Regenerative Therapies, Berlin, Germany; Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany; Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Julius Wolff Institute, Berlin, Germany
| | - Timo Gaber
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany; Berlin Institute of Health at Charité Universitätsmedizin Berlin, Center for Regenerative Therapies, Berlin, Germany.
| | - Frank Buttgereit
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany; Berlin Institute of Health at Charité Universitätsmedizin Berlin, Center for Regenerative Therapies, Berlin, Germany
| |
Collapse
|
36
|
Abstract
Fracture healing is a complex, multistep process that is highly sensitive to mechanical signaling. To optimize repair, surgeons prescribe immediate weight-bearing as-tolerated within 24 hours after surgical fixation; however, this recommendation is based on anecdotal evidence and assessment of bulk healing outcomes (e.g., callus size, bone volume, etc.). Given challenges in accurately characterizing the mechanical environment and the ever-changing properties of the regenerate, the principles governing mechanical regulation of repair, including their cell and molecular basis, are not yet well defined. However, the use of mechanobiological rodent models, and their relatively large genetic toolbox, combined with recent advances in imaging approaches and single-cell analyses is improving our understanding of the bone microenvironment in response to loading. This review describes the identification and characterization of distinct cell populations involved in bone healing and highlights the most recent findings on mechanical regulation of bone homeostasis and repair with an emphasis on osteo-angio coupling. A discussion on aging and its impact on bone mechanoresponsiveness emphasizes the need for novel mechanotherapeutics that can re-sensitize skeletal stem and progenitor cells to physical rehabilitation protocols.
Collapse
Affiliation(s)
- Tareq Anani
- Department of Orthopedic Surgery, New York University Langone Health, New York, NY 10010, USA
| | - Alesha B Castillo
- Department of Orthopedic Surgery, New York University Langone Health, New York, NY 10010, USA; Department of Biomedical Engineering, Tandon School of Engineering, New York University, New York, NY 11201, USA; Department of Veterans Affairs, New York Harbor Healthcare System, Manhattan Campus, New York, NY 10010, USA.
| |
Collapse
|
37
|
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: 20] [Impact Index Per Article: 5.0] [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
|
38
|
Wolter A, Rapp AE, Durst MS, Hildebrand L, Löhning M, Buttgereit F, Schmidt-Bleek K, Jirkof P, Lang A. Systematic review on the reporting accuracy of experimental details in publications using mouse femoral fracture models. Bone 2021; 152:116088. [PMID: 34175502 DOI: 10.1016/j.bone.2021.116088] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 11/28/2022]
Abstract
The outcomes of animal experiments can be influenced by a variety of factors. Thus, precise reporting is necessary to provide reliable and reproducible data. Initiatives such as the ARRIVE guidelines have been enrolled during the last decade to provide a road map for sufficient reporting. To understand the sophisticated process of bone regeneration and to develop new therapeutic strategies, small rodents, especially mice, are frequently used in bone healing research. Since many factors might influence the results from those studies, we performed a systematic literature search from 2010 to 2019 to identify studies involving mouse femoral fracture models (stable fixation) and evaluated the reporting of general and model-specific experimental details. 254 pre-selected publications were systematically analyzed, showing a high reporting accuracy for the used mouse strain, the age or developmental stage and sex of mice as well as model-specific information on fixation methods and fracturing procedures. However, reporting was more often insufficient in terms of mouse substrains and genetic backgrounds of genetically modified mice, body weight, hygiene monitoring/immune status of the animal, anesthesia, and analgesia. Consistent and reliable reporting of experimental variables in mouse fracture surgeries will improve scientific quality, enhance animal welfare, and foster translation into the clinic.
Collapse
Affiliation(s)
- Angelique Wolter
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, Pitzer Laboratory of Osteoarthritis Research, Berlin, Germany.
| | - Anna E Rapp
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, Pitzer Laboratory of Osteoarthritis Research, Berlin, Germany.
| | - Mattea S Durst
- Division of Surgical Research, University Hospital Zurich, University Zurich, Switzerland.
| | - Laura Hildebrand
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Center for Regenerative Therapies, Berlin, Germany.
| | - Max Löhning
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, Pitzer Laboratory of Osteoarthritis Research, Berlin, Germany.
| | - Frank Buttgereit
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, Pitzer Laboratory of Osteoarthritis Research, Berlin, Germany.
| | - Katharina Schmidt-Bleek
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Center for Regenerative Therapies, Berlin, Germany; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Julius Wolff Institute, Berlin, Germany.
| | - Paulin Jirkof
- Division of Surgical Research, University Hospital Zurich, University Zurich, Switzerland; Office for Animal Welfare and 3Rs, University of Zurich, Switzerland.
| | - Annemarie Lang
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, Pitzer Laboratory of Osteoarthritis Research, Berlin, Germany.
| |
Collapse
|
39
|
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: 171] [Impact Index Per Article: 42.8] [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
|
40
|
Fracture Healing Research-Shift towards In Vitro Modeling? Biomedicines 2021; 9:biomedicines9070748. [PMID: 34203470 PMCID: PMC8301383 DOI: 10.3390/biomedicines9070748] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/18/2021] [Accepted: 06/23/2021] [Indexed: 01/07/2023] Open
Abstract
Fractures are one of the most frequently occurring traumatic events worldwide. Approximately 10% of fractures lead to bone healing disorders, resulting in strain for affected patients and enormous costs for society. In order to shed light into underlying mechanisms of bone regeneration (habitual or disturbed), and to develop new therapeutic strategies, various in vivo, ex vivo and in vitro models can be applied. Undeniably, in vivo models include the systemic and biological situation. However, transferability towards the human patient along with ethical concerns regarding in vivo models have to be considered. Fostered by enormous technical improvements, such as bioreactors, on-a-chip-technologies and bone tissue engineering, sophisticated in vitro models are of rising interest. These models offer the possibility to use human cells from individual donors, complex cell systems and 3D models, therefore bridging the transferability gap, providing a platform for the introduction of personalized precision medicine and finally sparing animals. Facing diverse processes during fracture healing and thus various scientific opportunities, the reliability of results oftentimes depends on the choice of an appropriate model. Hence, we here focus on categorizing available models with respect to the requirements of the scientific approach.
Collapse
|
41
|
Tucci MA, Pride Y, Strickland S, Marocho SMS, Jackson RJ, Jefferson JR, Chade AR, Grill RJ, Grayson BE. Delayed Systemic Treatment with Cannabinoid Receptor 2 Agonist Mitigates Spinal Cord Injury-Induced Osteoporosis More Than Acute Treatment Directly after Injury. Neurotrauma Rep 2021; 2:270-284. [PMID: 34223557 PMCID: PMC8244511 DOI: 10.1089/neur.2020.0059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Nearly all persons with spinal cord injury (SCI) will develop osteoporosis following injury, and further, up to 50% of all persons with SCI will sustain a fracture during their lives. The unique mechanisms driving osteoporosis following SCI remain unknown. The cannabinoid system modulation of bone metabolism through cannabinoid 1/2 (CB1/2) has been of increasing interest for the preservation of bone mass and density in models of osteoporosis. Using a thoracic vertebral level 8 (T8) complete transection in a mouse model, we performed daily treatment with a selective CB2 receptor agonist, HU308, compared with SCI-vehicle-treated and naïve control animals either immediately after injury for 40 days, or in a delayed paradigm, following 3 months after injury. The goal was to prevent or potentially reverse SCI-induced osteoporosis. In the acute phase, administration of the CB2 agonist was not able to preserve the rapid loss of cancellous bone. In the delayed-treatment paradigm, in cortical bone, HU308 increased cortical-area to total-area ratio and periosteal perimeter in the femur, and improved bone density in the distal femur and proximal tibia. Further, we report changes to the metaphyseal periosteum with increased presence of adipocyte and fat mass in the periosteum of SCI animals, which was not present in naïve animals. The layer of fat increased markedly in HU308-treated animals compared with SCI-vehicle-treated animals. Overall, these data show that CB2 agonism targets a number of cell types that can influence overall bone quality.
Collapse
Affiliation(s)
- Michelle A. Tucci
- Department of Anesthesiology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Yilianys Pride
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Suzanne Strickland
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Susanna M. Salazar Marocho
- Department of Biomedical Materials Science, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Ramon J. Jackson
- Department of Anesthesiology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Joshua R. Jefferson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Alejandro R. Chade
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Department of Radiology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Raymond J. Grill
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Bernadette E. Grayson
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
42
|
Carrasco E, Gómez de Las Heras MM, Gabandé-Rodríguez E, Desdín-Micó G, Aranda JF, Mittelbrunn M. The role of T cells in age-related diseases. Nat Rev Immunol 2021; 22:97-111. [PMID: 34099898 DOI: 10.1038/s41577-021-00557-4] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2021] [Indexed: 12/11/2022]
Abstract
Age-related T cell dysfunction can lead to failure of immune tolerance mechanisms, resulting in aberrant T cell-driven cytokine and cytotoxic responses that ultimately cause tissue damage. In this Review, we discuss the role of T cells in the onset and progression of age-associated conditions, focusing on cardiovascular disorders, metabolic dysfunction, neuroinflammation and defective tissue repair and regeneration. We present different mechanisms by which T cells contribute to inflammageing and might act as modulators of age-associated diseases, including through enhanced pro-inflammatory and cytotoxic activity, defective clearance of senescent cells or regulation of the gut microbiota. Finally, we propose that 'resetting' immune system tolerance or targeting pathogenic T cells could open up new therapeutic opportunities to boost resilience to age-related diseases.
Collapse
Affiliation(s)
- Elisa Carrasco
- Departamento de Biología, Facultad de Ciencias (UAM); Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Biología Molecular, Facultad de Ciencias (UAM); Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuel M Gómez de Las Heras
- Departamento de Biología Molecular, Facultad de Ciencias (UAM); Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Enrique Gabandé-Rodríguez
- Departamento de Biología Molecular, Facultad de Ciencias (UAM); Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Gabriela Desdín-Micó
- Departamento de Biología Molecular, Facultad de Ciencias (UAM); Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Juan Francisco Aranda
- Departamento de Biología Molecular, Facultad de Ciencias (UAM); Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Maria Mittelbrunn
- Departamento de Biología Molecular, Facultad de Ciencias (UAM); Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain. .,Instituto de Investigación Sanitaria del Hospital 12 de Octubre (i+12), Madrid, Spain.
| |
Collapse
|
43
|
Wu P, Zhou J, Wu Y, Zhao L. The emerging role of Interleukin 37 in bone homeostasis and inflammatory bone diseases. Int Immunopharmacol 2021; 98:107803. [PMID: 34091255 DOI: 10.1016/j.intimp.2021.107803] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 02/05/2023]
Abstract
Interleukin 37 (IL-37) is a newly identified cytokine that belongs to the IL-1 family. Unlike other members of the IL-1 family, it has been demonstrated that IL-37 possesses anti-inflammatory characteristics in both innate and acquired immune responses. Recently, significant progress has been made in understanding the role of IL-37 in inflammatory signaling pathways. Meanwhile, IL-37 has also attracted more and more attention in bone homeostasis and inflammatory bone diseases. The latest studies have revealed that IL-37 palys an essential role in the regulation of osteoclastogenesis and osteoblastogenesis. The levels of IL-37 are abnormal in patients with inflammatory bone diseases such as rheumatoid arthritis (RA), osteoarthritis (OA), ankylosing spondylitis (AS), and periodontitis. In addition, in vivo studies have further confirmed that recombinant IL-37 treatment displayed therapeutic potential in these diseases. The present review article aims to provide an overview describing the biological functions of IL-37 in bone homeostasis and inflammatory bone diseases, thus shedding new light on a novel therapeutic strategy in the future.
Collapse
Affiliation(s)
- Peiyao Wu
- Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jieyu Zhou
- Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yafei Wu
- Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Lei Zhao
- Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
44
|
Sadowska JM, Ginebra MP. Inflammation and biomaterials: role of the immune response in bone regeneration by inorganic scaffolds. J Mater Chem B 2021; 8:9404-9427. [PMID: 32970087 DOI: 10.1039/d0tb01379j] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The regulatory role of the immune system in maintaining bone homeostasis and restoring its functionality, when disturbed due to trauma or injury, has become evident in recent years. The polarization of macrophages, one of the main constituents of the immune system, into the pro-inflammatory or anti-inflammatory phenotype has great repercussions for cellular crosstalk and the subsequent processes needed for proper bone regeneration such as angiogenesis and osteogenesis. In certain scenarios, the damaged osseous tissue requires the placement of synthetic bone grafts to facilitate the healing process. Inorganic biomaterials such as bioceramics or bioactive glasses are the most widely used due to their resemblance to the mineral phase of bone and superior osteogenic properties. The immune response of the host to the inorganic biomaterial, which is of an exogenous nature, might determine its fate, leading either to active bone regeneration or its failure. Therefore, various strategies have been employed, like the modification of structural/chemical features or the incorporation of bioactive molecules, to tune the interplay with the immune cells. Understanding how these particular modifications impact the polarization of macrophages and further osteogenic and osteoclastogenic events is of great interest in view of designing a new generation of osteoimmunomodulatory materials that support the regeneration of osseous tissue during all stages of bone healing.
Collapse
Affiliation(s)
- Joanna M Sadowska
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), Ireland
| | - Maria-Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya, Av. Eduard Maristany 16, 08019 Barcelona, Spain. and Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
| |
Collapse
|
45
|
Negrescu AM, Cimpean A. The State of the Art and Prospects for Osteoimmunomodulatory Biomaterials. MATERIALS (BASEL, SWITZERLAND) 2021; 14:1357. [PMID: 33799681 PMCID: PMC7999637 DOI: 10.3390/ma14061357] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/04/2021] [Accepted: 03/08/2021] [Indexed: 12/14/2022]
Abstract
The critical role of the immune system in host defense against foreign bodies and pathogens has been long recognized. With the introduction of a new field of research called osteoimmunology, the crosstalk between the immune and bone-forming cells has been studied more thoroughly, leading to the conclusion that the two systems are intimately connected through various cytokines, signaling molecules, transcription factors and receptors. The host immune reaction triggered by biomaterial implantation determines the in vivo fate of the implant, either in new bone formation or in fibrous tissue encapsulation. The traditional biomaterial design consisted in fabricating inert biomaterials capable of stimulating osteogenesis; however, inconsistencies between the in vitro and in vivo results were reported. This led to a shift in the development of biomaterials towards implants with osteoimmunomodulatory properties. By endowing the orthopedic biomaterials with favorable osteoimmunomodulatory properties, a desired immune response can be triggered in order to obtain a proper bone regeneration process. In this context, various approaches, such as the modification of chemical/structural characteristics or the incorporation of bioactive molecules, have been employed in order to modulate the crosstalk with the immune cells. The current review provides an overview of recent developments in such applied strategies.
Collapse
Affiliation(s)
| | - Anisoara Cimpean
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania;
| |
Collapse
|
46
|
Knecht RS, Bucher CH, Van Linthout S, Tschöpe C, Schmidt-Bleek K, Duda GN. Mechanobiological Principles Influence the Immune Response in Regeneration: Implications for Bone Healing. Front Bioeng Biotechnol 2021; 9:614508. [PMID: 33644014 PMCID: PMC7907627 DOI: 10.3389/fbioe.2021.614508] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 01/22/2021] [Indexed: 12/13/2022] Open
Abstract
A misdirected or imbalanced local immune composition is often one of the reasons for unsuccessful regeneration resulting in scarring or fibrosis. Successful healing requires a balanced initiation and a timely down-regulation of the inflammation for the re-establishment of a biologically and mechanically homeostasis. While biomaterial-based approaches to control local immune responses are emerging as potential new treatment options, the extent to which biophysical material properties themselves play a role in modulating a local immune niche response has so far been considered only occasionally. The communication loop between extracellular matrix, non-hematopoietic cells, and immune cells seems to be specifically sensitive to mechanical cues and appears to play a role in the initiation and promotion of a local inflammatory setting. In this review, we focus on the crosstalk between ECM and its mechanical triggers and how they impact immune cells and non-hematopoietic cells and their crosstalk during tissue regeneration. We realized that especially mechanosensitive receptors such as TRPV4 and PIEZO1 and the mechanosensitive transcription factor YAP/TAZ are essential to regeneration in various organ settings. This indicates novel opportunities for therapeutic approaches to improve tissue regeneration, based on the immune-mechanical principles found in bone but also lung, heart, and skin.
Collapse
Affiliation(s)
- Raphael S Knecht
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christian H Bucher
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sophie Van Linthout
- Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Carsten Tschöpe
- Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany.,Department of Cardiology, Charite'-Universitätsmedizin Berlin, Berlin, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Georg N Duda
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
47
|
Spatio-Temporal Bone Remodeling after Hematopoietic Stem Cell Transplantation. Int J Mol Sci 2020; 22:ijms22010267. [PMID: 33383915 PMCID: PMC7795370 DOI: 10.3390/ijms22010267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/17/2020] [Accepted: 12/23/2020] [Indexed: 12/28/2022] Open
Abstract
The interaction of hematopoietic cells and the bone microenvironment to maintain bone homeostasis is increasingly appreciated. We hypothesized that the transfer of allogeneic T lymphocytes has extensive effects on bone biology and investigated trabecular and cortical bone structures, the osteoblast reconstitution, and the bone vasculature in experimental hematopoietic stem cell transplantations (HSCT). Allogeneic or syngeneic hematopoietic stem cells (HSC) and allogeneic T lymphocytes were isolated and transferred in a murine model. After 20, 40, and 60 days, bone structures were visualized using microCT and histology. Immune cells were monitored using flow cytometry and bone vessels, bone cells and immune cells were fluorescently stained and visualized. Remodeling of the bone substance, the bone vasculature and bone cell subsets were found to occur as early as day +20 after allogeneic HSCT (including allogeneic T lymphocytes) but not after syngeneic HSCT. We discovered that allogeneic HSCT (including allogeneic T lymphocytes) results in a transient increase of trabecular bone number and bone vessel density. This was paralleled by a cortical thinning as well as disruptive osteoblast lining and loss of B lymphocytes. In summary, our data demonstrate that the adoptive transfer of allogeneic HSCs and allogeneic T lymphocytes can induce profound structural and spatial changes of bone tissue homeostasis as well as bone marrow cell composition, underlining the importance of the adaptive immune system for maintaining a balanced bone biology.
Collapse
|
48
|
Longitudinal time-lapse in vivo micro-CT reveals differential patterns of peri-implant bone changes after subclinical bacterial infection in a rat model. Sci Rep 2020; 10:20901. [PMID: 33262377 PMCID: PMC7708479 DOI: 10.1038/s41598-020-77770-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 11/17/2020] [Indexed: 02/06/2023] Open
Abstract
Subclinical infection associated with orthopedic devices can be challenging to diagnose. The goal of this study was to evaluate longitudinal, microcomputed tomography (microCT) imaging in a rat model of subclinical orthopedic device-related infection caused by Staphylococcus epidermidis and four different Cutibacterium (previously Propionibacterium) acnes strains, and compare outcomes with non-inoculated and historical S. aureus-inoculated controls. Sterile screws or screws colonized with bacteria were placed in the tibia of 38 adult Wistar rats [n = 6 sterile screws; n = 6 S. epidermidis-colonized screws; n = 26 C. acnes-colonized screws (covering all three main subspecies)]. Regular microCT scans were taken over 28 days and processed for quantitative time-lapse imaging with dynamic histomorphometry. At euthanasia, tissues were processed for semiquantitative histopathology or quantitative bacteriology. All rats receiving sterile screws were culture-negative at euthanasia and displayed progressive bony encapsulation of the screw. All rats inoculated with S. epidermidis-colonized screws were culture-positive and displayed minor changes in peri-implant bone, characteristic of subclinical infection. Five of the 17 rats in the C. acnes inoculated group were culture positive at euthanasia and displayed bone changes at the interface of the screw and bone, but not deeper in the peri-implant bone. Dynamic histomorphometry revealed significant differences in osseointegration, bone remodeling and periosteal reactions between groups that were not measurable by visual observation of still microCT images. Our study illustrates the added value of merging 3D microCT data from subsequent timepoints and producing inherently richer 4D data for the detection and characterization of subclinical orthopedic infections, whilst also reducing animal use.
Collapse
|
49
|
Abstract
Inflammatory cardiomyopathy, characterized by inflammatory cell infiltration into the myocardium and a high risk of deteriorating cardiac function, has a heterogeneous aetiology. Inflammatory cardiomyopathy is predominantly mediated by viral infection, but can also be induced by bacterial, protozoal or fungal infections as well as a wide variety of toxic substances and drugs and systemic immune-mediated diseases. Despite extensive research, inflammatory cardiomyopathy complicated by left ventricular dysfunction, heart failure or arrhythmia is associated with a poor prognosis. At present, the reason why some patients recover without residual myocardial injury whereas others develop dilated cardiomyopathy is unclear. The relative roles of the pathogen, host genomics and environmental factors in disease progression and healing are still under discussion, including which viruses are active inducers and which are only bystanders. As a consequence, treatment strategies are not well established. In this Review, we summarize and evaluate the available evidence on the pathogenesis, diagnosis and treatment of myocarditis and inflammatory cardiomyopathy, with a special focus on virus-induced and virus-associated myocarditis. Furthermore, we identify knowledge gaps, appraise the available experimental models and propose future directions for the field. The current knowledge and open questions regarding the cardiovascular effects associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are also discussed. This Review is the result of scientific cooperation of members of the Heart Failure Association of the ESC, the Heart Failure Society of America and the Japanese Heart Failure Society.
Collapse
|
50
|
Berkmann JC, Herrera Martin AX, Pontremoli C, Zheng K, Bucher CH, Ellinghaus A, Boccaccini AR, Fiorilli S, Vitale Brovarone C, Duda GN, Schmidt-Bleek K. In Vivo Validation of Spray-Dried Mesoporous Bioactive Glass Microspheres Acting as Prolonged Local Release Systems for BMP-2 to Support Bone Regeneration. Pharmaceutics 2020; 12:pharmaceutics12090823. [PMID: 32872353 PMCID: PMC7559713 DOI: 10.3390/pharmaceutics12090823] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 08/24/2020] [Accepted: 08/26/2020] [Indexed: 12/30/2022] Open
Abstract
Bone morphogenetic protein-2 (BMP-2) is a known key mediator of physiological bone regeneration and is clinically approved for selected musculoskeletal interventions. Yet, broad usage of this growth factor is impeded due to side effects that are majorly evoked by high dosages and burst release kinetics. In this study, mesoporous bioactive glass microspheres (MBGs), produced by an aerosol-assisted spray-drying scalable process, were loaded with BMP-2 resulting in prolonged, low-dose BMP-2 release without affecting the material characteristics. In vitro, MBGs were found to be cytocompatible and to induce a pro-osteogenic response in primary human mesenchymal stromal cells (MSCs). In a pre-clinical rodent model, BMP-2 loaded MBGs significantly enhanced bone formation and influenced the microarchitecture of newly formed bone. The MBG carriers alone performed equal to the untreated (empty) control in most parameters tested, while additionally exerting mild pro-angiogenic effects. Using MBGs as a biocompatible, pro-regenerative carrier for local and sustained low dose BMP-2 release could limit side effects, thus enabling a safer usage of BMP-2 as a potent pro-osteogenic growth factor.
Collapse
Affiliation(s)
- Julia C. Berkmann
- Julius-Wolff-Institut, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, 13353 Berlin, Germany; (J.C.B.); (A.X.H.M.); (C.H.B.); (G.N.D.)
- Berlin-Brandenburg School for Regenerative Therapies, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Aaron X. Herrera Martin
- Julius-Wolff-Institut, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, 13353 Berlin, Germany; (J.C.B.); (A.X.H.M.); (C.H.B.); (G.N.D.)
- Berlin-Brandenburg School for Regenerative Therapies, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Carlotta Pontremoli
- Department of Applied Science and Technology, Politecnico di Torino, 10129 Turin, Italy; (C.P.); (S.F.); (C.V.B.)
| | - Kai Zheng
- Institute of Biomaterials, University of Erlangen-Nuremberg, 91058 Erlangen, Germany; (K.Z.); (A.R.B.)
| | - Christian H. Bucher
- Julius-Wolff-Institut, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, 13353 Berlin, Germany; (J.C.B.); (A.X.H.M.); (C.H.B.); (G.N.D.)
- Berlin-Brandenburg School for Regenerative Therapies, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
- BIH Center for Regenerative Therapies, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, 13353 Berlin, Germany;
| | - Agnes Ellinghaus
- BIH Center for Regenerative Therapies, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, 13353 Berlin, Germany;
| | - Aldo R. Boccaccini
- Institute of Biomaterials, University of Erlangen-Nuremberg, 91058 Erlangen, Germany; (K.Z.); (A.R.B.)
| | - Sonia Fiorilli
- Department of Applied Science and Technology, Politecnico di Torino, 10129 Turin, Italy; (C.P.); (S.F.); (C.V.B.)
| | - Chiara Vitale Brovarone
- Department of Applied Science and Technology, Politecnico di Torino, 10129 Turin, Italy; (C.P.); (S.F.); (C.V.B.)
| | - Georg N. Duda
- Julius-Wolff-Institut, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, 13353 Berlin, Germany; (J.C.B.); (A.X.H.M.); (C.H.B.); (G.N.D.)
- BIH Center for Regenerative Therapies, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, 13353 Berlin, Germany;
| | - Katharina Schmidt-Bleek
- Julius-Wolff-Institut, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, 13353 Berlin, Germany; (J.C.B.); (A.X.H.M.); (C.H.B.); (G.N.D.)
- BIH Center for Regenerative Therapies, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, 13353 Berlin, Germany;
- Correspondence: ; Tel.: +49-(0)30-450659209
| |
Collapse
|