1
|
Horenberg AL, Ren Y, Zeng EZ, Rindone AN, Pathak AP, Grayson WL. 3D imaging reveals changes in the neurovascular architecture of the murine calvarium with aging. Bone Res 2025; 13:24. [PMID: 39984434 PMCID: PMC11845787 DOI: 10.1038/s41413-025-00401-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 12/18/2024] [Accepted: 12/31/2024] [Indexed: 02/23/2025] Open
Abstract
Calvarial nerves, along with vasculature, influence skull formation during development and following injury, but it remains unclear how calvarial nerves are spatially distributed during postnatal growth and aging. Studying the spatial distribution of nerves in the skull remains a challenge due to a lack of methods to quantify 3D structures in intact bone. To visualize calvarial 3D neurovascular architecture, we imaged nerves and endothelial cells with lightsheet microscopy. We employed machine-learning-based segmentation to facilitate high-resolution characterization from post-natal day 0 (P0) to 80 weeks. We found that TUBB3+ nerve density decreased with aging with the frontal bone demonstrating earlier onset age-related nerve loss than the parietal bone. In addition, nerves in the periosteum and dura mater exhibited similar yet distinct temporal patterns of nerve growth and loss. While no difference was observed in TUBB3+ nerves during skeletal maturation (P0 → 12 weeks), we did observe an increase in the volume of unmyelinated nerves in the dura mater. Regarding calvarial vasculature, larger CD31hiEmcn- vessel fraction increased with aging, while CD31hiEmcnhi vessel fraction was reduced. Throughout all ages, calvarial nerves maintained a preferential spatial association with CD31hiEmcnhi vessels, however, this association decreased with aging. Additionally, we used a model of Apert syndrome to explore the impact of suture-related disease on neurovascular architecture. Collectively, this 3D, spatiotemporal characterization of calvarial nerves throughout the lifespan and provides new insights into age-induced neurovascular architecture.
Collapse
Affiliation(s)
- Allison L Horenberg
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yunke Ren
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eric Z Zeng
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexandra N Rindone
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Arvind P Pathak
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Sciences, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Electrical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Warren L Grayson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
2
|
Perepletchikova D, Kuchur P, Basovich L, Khvorova I, Lobov A, Azarkina K, Aksenov N, Bozhkova S, Karelkin V, Malashicheva A. Endothelial-mesenchymal crosstalk drives osteogenic differentiation of human osteoblasts through Notch signaling. Cell Commun Signal 2025; 23:100. [PMID: 39972367 PMCID: PMC11841332 DOI: 10.1186/s12964-025-02096-0] [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: 11/28/2024] [Accepted: 02/08/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Angiogenesis and osteogenesis are closely interrelated. The interaction between endothelial and bone-forming cells, such as osteoblasts, is crucial for normal bone development and repair. Juxtacrine and paracrine mechanisms play key roles in cell differentiation towards the osteogenic direction, assuming the direct effect of endothelium on osteogenic differentiation. However, the mechanisms of this interplay have yet to be thoroughly studied. METHODS Isolated endothelial cells (EC) from human umbilical vein and human osteoblasts (OB) from the epiphysis of the femur or tibia were cultured in direct and indirect (separated by membrane) contact in vitro under the osteogenic differentiation conditions. Osteogenic differentiation was verified by RT-PCR, and alizarin red staining. Shotgun proteomics and RNA-sequencing were used to compare both EC and OB under different co-culture conditions to assess the mechanisms of EC-OB interplay. To verify the role of Notch signaling, experiments with Notch modulation in EC were performed by EC lentiviral transduction with further co-cultivation with OB. Additionally, the effect of Notch modulation in EC was assessed by RNA-sequencing. RESULTS EC have opposite effects on osteogenic differentiation depending on the co-culture conditions with OB. In direct contact, EC enhance osteogenic differentiation, but in indirect cultures, EC suppress it. Our proteotranscriptomic analysis revealed that the osteosuppressive effect is related to the action of paracrine factors secreted by EC, while the osteoinductive properties of EC are mediated by the Notch signaling pathway, which can be activated only upon a physical contact of EC with OB. Indeed, in the direct co-culture, the knockdown of Notch1 and Notch3 receptors in EC has an inhibitory effect on the OB osteogenic differentiation, whereas activation of Notch by intracellular domain of either Notch1 or Notch3 in EC has an inductive effect on the OB osteogenic differentiation. CONCLUSION The data indicate the dual role of the endothelium in regulating osteogenic differentiation and highlight the unique role of the Notch signaling pathway in inducing osteogenic differentiation during cell-to-cell interactions. The findings of the study emphasize the importance of intercellular communication in the regulation of osteoblast differentiation during bone development and maintenance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Svetlana Bozhkova
- Vreden National Medical Research Center of Traumatology and Orthopedics, Saint- Petersburg, Russia
| | - Vitaliy Karelkin
- Vreden National Medical Research Center of Traumatology and Orthopedics, Saint- Petersburg, Russia
| | | |
Collapse
|
3
|
Perepletchikova D, Malashicheva A. Communication between endothelial cells and osteoblasts in regulation of bone homeostasis: Notch players. Stem Cell Res Ther 2025; 16:56. [PMID: 39920854 PMCID: PMC11806792 DOI: 10.1186/s13287-025-04176-x] [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: 07/25/2024] [Accepted: 01/23/2025] [Indexed: 02/09/2025] Open
Abstract
Endothelial cells coat blood vessels and release molecular signals to affect the fate of other cells. Endothelial cells can adjust their behavior in response to the changing microenvironmental conditions. During bone regeneration, bone tissue cells release factors that promote blood vessel growth. Notch is a key signaling that regulates cell fate decisions in many tissues and plays an important role in bone tissue development and homeostasis. Understanding the interplay between angiogenesis and osteogenesis is currently a focus of research efforts in order to facilitate and improve osteogenesis when needed. Our review explores the cellular and molecular mechanisms including Notch-dependent endothelial-MSC communication that drive osteogenesis-angiogenesis processes and their effects on bone remodeling and repair.
Collapse
Affiliation(s)
| | - Anna Malashicheva
- Institute of Cytology Russian Academy of Science, St. Petersburg, Russia, 194064.
| |
Collapse
|
4
|
Bertolucci V, Ninomiya AF, Longato GB, Kaneko LO, Nonose N, Scariot PPM, Messias LHD. Bioactive Compounds from Propolis on Bone Homeostasis: A Narrative Review. Antioxidants (Basel) 2025; 14:81. [PMID: 39857415 PMCID: PMC11762496 DOI: 10.3390/antiox14010081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
This narrative review explores the potential effects of Propolis and its bioactive compounds on bone health. Propolis, a resinous product collected by bees, is renowned for its antimicrobial, anti-inflammatory, and antioxidant properties. Recent research emphasizes its positive role in osteogenesis, primarily through the modulation of osteoclast and osteoblast activity via molecular pathways. Key mechanisms include reducing inflammatory cytokines, protecting against oxidative stress, and upregulating growth factor essential for bone formation. While compounds such as Caffeic Acid Phenethyl Ester, Apigenin, Quercetin, and Ferulic Acid have been well-documented, emerging evidence points to the significant roles of less-studied compounds like Pinocembrin, Kaempferol, p-Coumaric acid, and Galangin. This review synthesizes the current literature, focusing on the mechanisms by which these bioactive compounds influence osteogenesis. Firstly, it explores the techniques for characterizing bioactive compounds presented in propolis, the chemogeographic variations in its composition, and the effects of both crude extracts and isolated compounds on bone tissue, offering a comprehensive analysis of recent findings across different experimental models. Further, it discusses the effects of Propolis compounds on bone health. In summary, these compounds modulate signaling pathways, including nuclear factor kappa beta, wingless-related integration site, mitogen-activated protein kinase, vascular endothelial growth factor, and reactive oxygen species. These pathways influence the receptor activator of nuclear factor kappa-β/receptor activator of nuclear factor kappa-β ligand/osteoprotegerin system, fostering bone cell differentiation. This regulation mitigates excessive osteoclast formation, stimulates osteoblast activity, and ultimately contributes to the restoration of bone homeostasis by maintaining a balanced bone remodeling process.
Collapse
Affiliation(s)
- Vanessa Bertolucci
- Research Group on Technology Applied to Exercise Physiology—GTAFE, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil; (V.B.); (N.N.)
| | - André Felipe Ninomiya
- Research Group on Technology Applied to Exercise Physiology—GTAFE, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil; (V.B.); (N.N.)
| | - Giovanna Barbarini Longato
- Research Laboratory in Molecular Pharmacology of Bioactive Compounds, São Francisco University, Bragança Paulista 12916-900, SP, Brazil;
| | - Luisa Oliveira Kaneko
- Research Group on Technology Applied to Exercise Physiology—GTAFE, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil; (V.B.); (N.N.)
| | - Nilson Nonose
- Research Group on Technology Applied to Exercise Physiology—GTAFE, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil; (V.B.); (N.N.)
| | - Pedro Paulo Menezes Scariot
- Research Group on Technology Applied to Exercise Physiology—GTAFE, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil; (V.B.); (N.N.)
| | - Leonardo Henrique Dalcheco Messias
- Research Group on Technology Applied to Exercise Physiology—GTAFE, Health Sciences Postgraduate Program, São Francisco University, Bragança Paulista 12916-900, SP, Brazil; (V.B.); (N.N.)
| |
Collapse
|
5
|
Cain TL, Derecka M, McKinney-Freeman S. The role of the haematopoietic stem cell niche in development and ageing. Nat Rev Mol Cell Biol 2025; 26:32-50. [PMID: 39256623 DOI: 10.1038/s41580-024-00770-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2024] [Indexed: 09/12/2024]
Abstract
Blood production depends on rare haematopoietic stem cells (HSCs) and haematopoietic stem and progenitor cells (HSPCs) that ultimately take up residence in the bone marrow during development. HSPCs and HSCs are subject to extrinsic regulation by the bone marrow microenvironment, or niche. Studying the interactions between HSCs and their niche is critical for improving ex vivo culturing conditions and genetic manipulation of HSCs, which is pivotal for improving autologous HSC therapies and transplantations. Additionally, understanding how the complex molecular network in the bone marrow is altered during ageing is paramount for developing novel therapeutics for ageing-related haematopoietic disorders. HSCs are unique amongst stem and progenitor cell pools in that they engage with multiple physically distinct niches during their ontogeny. HSCs are specified from haemogenic endothelium in the aorta, migrate to the fetal liver and, ultimately, colonize their final niche in the bone marrow. Recent studies employing single-cell transcriptomics and microscopy have identified novel cellular interactions that govern HSC specification and engagement with their niches throughout ontogeny. New lineage-tracing models and microscopy tools have raised questions about the numbers of HSCs specified, as well as the functional consequences of HSCs interacting with each developmental niche. Advances have also been made in understanding how these niches are modified and perturbed during ageing, and the role of these altered interactions in haematopoietic diseases. In this Review, we discuss these new findings and highlight the questions that remain to be explored.
Collapse
Affiliation(s)
- Terri L Cain
- Department of Haematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Marta Derecka
- Department of Haematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | |
Collapse
|
6
|
Xv D, Cao Y, Hou Y, Hu Y, Li M, Xie C, Lu X. Polyphenols and Functionalized Hydrogels for Osteoporotic Bone Regeneration. Macromol Rapid Commun 2025; 46:e2400653. [PMID: 39588839 DOI: 10.1002/marc.202400653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/02/2024] [Indexed: 11/27/2024]
Abstract
Osteoporosis induces severe oxidative stress and disrupts bone metabolism, complicating the treatment of bone defects. Current therapies often have side effects and require lengthy bone regeneration periods. Hydrogels, known for their flexible mechanical properties and degradability, are promising carriers for drugs and bioactive factors in bone tissue engineering. However, they lack the ability to regulate the local pathological environment of osteoporosis and expedite bone repair. Polyphenols, with antioxidative, anti-inflammatory, and bone metabolism-regulating properties, have emerged as a solution. Combining hydrogels and polyphenols, polyphenol-based hydrogels can regulate local bone metabolism and oxidative stress while providing mechanical support and tissue adhesion, promoting osteoporotic bone regeneration. This review first provides a brief overview of the types of polyphenols and the mechanisms of polyphenols in facilitating adhesion, antioxidant, anti-inflammatory, and bone metabolism modulation in modulating the pathological environment of osteoporosis. Next, this review examines recent advances in hydrogels for the treatment of osteoporotic bone defects, including their use in angiogenesis, oxidative stress modulation, drug delivery, and stem cell therapy. Finally, it highlights the latest research on polyphenol hydrogels in osteoporotic bone defect regeneration. Overall, this review aims to facilitate the clinical application of polyphenol hydrogels for the treatment of osteoporotic bone defects.
Collapse
Affiliation(s)
- Dejia Xv
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yuming Cao
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Yue Hou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Yuelin Hu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Minqi Li
- Department of Bone Metabolism, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250000, China
- Center of Osteoporosis and Bone Mineral Research, Shandong University, Jinan, 250000, China
| | - Chaoming Xie
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Xiong Lu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| |
Collapse
|
7
|
Jackett KN, Browne AT, Aber ER, Clements M, Kaplan RN. How the bone microenvironment shapes the pre-metastatic niche and metastasis. NATURE CANCER 2024; 5:1800-1814. [PMID: 39672975 DOI: 10.1038/s43018-024-00854-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/04/2024] [Indexed: 12/15/2024]
Abstract
The bone is a frequent metastatic site, with changes in the mineralized bone and the bone marrow milieu that can also prime other sites for metastasis by educating progenitor cells to support metastatic spread. Stromal and immune populations cooperatively maintain the organizationally complex bone niches and are dysregulated in the presence of a distant primary tumor and metastatic disease. Interrogating the bone niches that facilitate metastatic spread using innovative technologies holds the potential to aid in preventing metastasis in and mediated by the bone. Here, we review recent advances in bone niche biology and its adaptations in the context of cancer.
Collapse
Affiliation(s)
- Kailey N Jackett
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alice T Browne
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Etan R Aber
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Miranda Clements
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rosandra N Kaplan
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
8
|
Qi L, Hong S, Zhao T, Yan J, Ge W, Wang J, Fang X, Jiang W, Shen SG, Zhang L. DNA Tetrahedron Delivering miR-21-5p Promotes Senescent Bone Defects Repair through Synergistic Regulation of Osteogenesis and Angiogenesis. Adv Healthc Mater 2024; 13:e2401275. [PMID: 38979868 DOI: 10.1002/adhm.202401275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 06/08/2024] [Indexed: 07/10/2024]
Abstract
Compromised osteogenesis and angiogenesis is the character of stem cell senescence, which brought difficulties for bone defects repairing in senescent microenvironment. As the most abundant bone-related miRNA, miRNA-21-5p plays a crucial role in inducing osteogenic and angiogenic differentiation. However, highly efficient miR-21-5p delivery still confronts challenges including poor cellular uptake and easy degradation. Herein, TDN-miR-21-5p nanocomplex is constructed based on DNA tetrahedral (TDN) and has great potential in promoting osteogenesis and alleviating senescence of senescent bone marrow stem cells (O-BMSCs), simultaneously enhancing angiogenic capacity of senescent endothelial progenitor cells (O-EPCs). Of note, the activation of AKT and Erk signaling pathway may direct regulatory mechanism of TDN-miR-21-5p mediated osteogenesis and senescence of O-BMSCs. Also, TDN-miR-21-5p can indirectly mediate osteogenesis and senescence of O-BMSCs through pro-angiogenic growth factors secreted from O-EPCs. In addition, gelatin methacryloyl (GelMA) hydrogels are mixed with TDN and TDN-miR-21-5p to fabricate delivery scaffolds. TDN-miR-21-5p@GelMA scaffold exhibits greater bone repair with increased expression of osteogenic- and angiogenic-related markers in senescent critical-size cranial defects in vivo. Collectively, TDN-miR-21-5p can alleviate senescence and induce osteogenesis and angiogenesis in senescent microenvironment, which provides a novel candidate strategy for senescent bone repair and widen clinical application of TDNs-based gene therapy.
Collapse
Affiliation(s)
- Lei Qi
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Shebin Hong
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Tong Zhao
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Jinge Yan
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Weiwen Ge
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Jing Wang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Xin Fang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Weidong Jiang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Steve Gf Shen
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Lei Zhang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| |
Collapse
|
9
|
Reusswig F, An O, Deppermann C. Platelet life cycle during aging: function, production and clearance. Platelets 2024; 35:2433750. [PMID: 39618096 DOI: 10.1080/09537104.2024.2433750] [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: 09/04/2024] [Revised: 10/10/2024] [Accepted: 11/15/2024] [Indexed: 12/13/2024]
Abstract
Platelets are important players in hemostasis. Alterations in platelet number and/or function lead to life-threatening conditions like thrombosis, myocardial infarction and stroke. During aging, changes at the cellular, organ and systemic level occur that affect platelet counts, platelet functionality, the expression of platelet surface receptors, clearance markers as well as their interactions with immune cells. Understanding how these changes influence platelets can help to prevent the alterations of hemostasis and thrombosis we observe in the elderly. In this review, we highlight the respective changes at important sites of the platelet life cycle: bone marrow, liver and spleen, but also show how alterations in immunity contribute. We point out the necessity for further research on age-related systemic alterations in these systems and their interplay with platelets to better understand the complex processes that cause alterations in the platelet life cycle during aging.
Collapse
Affiliation(s)
- Friedrich Reusswig
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Olga An
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Carsten Deppermann
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Research Center for Immune Therapy, Forschungszentrum für Immuntherapie (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
10
|
Gao H, Nepovimova E, Adam V, Heger Z, Valko M, Wu Q, Kuca K. Age-associated changes in innate and adaptive immunity: role of the gut microbiota. Front Immunol 2024; 15:1421062. [PMID: 39351234 PMCID: PMC11439693 DOI: 10.3389/fimmu.2024.1421062] [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: 04/21/2024] [Accepted: 08/26/2024] [Indexed: 10/04/2024] Open
Abstract
Aging is generally regarded as an irreversible process, and its intricate relationship with the immune system has garnered significant attention due to its profound implications for the health and well-being of the aging population. As people age, a multitude of alterations occur within the immune system, affecting both innate and adaptive immunity. In the realm of innate immunity, aging brings about changes in the number and function of various immune cells, including neutrophils, monocytes, and macrophages. Additionally, certain immune pathways, like the cGAS-STING, become activated. These alterations can potentially result in telomere damage, the disruption of cytokine signaling, and impaired recognition of pathogens. The adaptive immune system, too, undergoes a myriad of changes as age advances. These include shifts in the number, frequency, subtype, and function of T cells and B cells. Furthermore, the human gut microbiota undergoes dynamic changes as a part of the aging process. Notably, the interplay between immune changes and gut microbiota highlights the gut's role in modulating immune responses and maintaining immune homeostasis. The gut microbiota of centenarians exhibits characteristics akin to those found in young individuals, setting it apart from the microbiota observed in typical elderly individuals. This review delves into the current understanding of how aging impacts the immune system and suggests potential strategies for reversing aging through interventions in immune factors.
Collapse
Affiliation(s)
- Haoyu Gao
- College of Life Science, Yangtze University, Jingzhou, China
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czechia
| | - Zbynek Heger
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czechia
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, Bratislava, Slovakia
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, China
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
- Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI), University of Granada, Granada, Spain
| |
Collapse
|
11
|
Debortoli C, Falguiere A, Campana F, Catherine JH, Tardivo D, Lan R. Utilization of a Cortical Xenogeneic Membrane for Guided Bone Regeneration: A Retrospective Case Series. J Clin Med 2024; 13:4575. [PMID: 39124840 PMCID: PMC11312857 DOI: 10.3390/jcm13154575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 07/22/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
Background: Guided bone regeneration (GBR) is a reliable technique used in vertical and horizontal bone defects. The posterior mandibular region is an area limited by anatomic constraints. The use of resorbable membranes with a cortical component could compensate for the lack of rigidity of resorbable membranes without the complications of non-resorbable membranes. The aim of this study was to evaluate the mean bone gains of a xenogeneic cortical membrane in horizontal and vertical bone defects in comparison with other membranes in the literature. Methods: A porcine cortical membrane was used to perform 7 GBR in the posterior mandibular region of five patients. Preoperative (T0) and six months postoperative (T1) cone beam computed tomography were superimposed to measure the horizontal and vertical bone gain. Implants were positioned at all sites, six months after GBR. Complications and bone resorption around the implants were also documented. Results: The mean horizontal and vertical bone gains were 3.83 ± 1.41 mm and 4.17 ± 1.86 mm, respectively. The analysis of repeatability was 0.997. As many as 40% of patients experienced pain refractory to analgesics. No exposure or infectious phenomenon was observed. Conclusions: This xenogeneic cortical membrane seemed to provide interesting results in the regeneration of horizontal and vertical bone defects. Comparative and prospective studies are necessary to validate the effectiveness of this membrane.
Collapse
Affiliation(s)
- Cyril Debortoli
- Oral Surgery Department, Assistance Publique des Hôpitaux de Marseille, 264 Avenue St Pierre, 13005 Marseille, France; (A.F.); (F.C.); (J.-H.C.); (R.L.)
| | - Arthur Falguiere
- Oral Surgery Department, Assistance Publique des Hôpitaux de Marseille, 264 Avenue St Pierre, 13005 Marseille, France; (A.F.); (F.C.); (J.-H.C.); (R.L.)
| | - Fabrice Campana
- Oral Surgery Department, Assistance Publique des Hôpitaux de Marseille, 264 Avenue St Pierre, 13005 Marseille, France; (A.F.); (F.C.); (J.-H.C.); (R.L.)
| | - Jean-Hugues Catherine
- Oral Surgery Department, Assistance Publique des Hôpitaux de Marseille, 264 Avenue St Pierre, 13005 Marseille, France; (A.F.); (F.C.); (J.-H.C.); (R.L.)
- Laboratory ISM, Aix-Marseille University, CNRS, EFS, 13005 Marseille, France
| | - Delphine Tardivo
- Laboratory ADES, Aix-Marseille University, CNRS, EFS, 13005 Marseille, France;
| | - Romain Lan
- Oral Surgery Department, Assistance Publique des Hôpitaux de Marseille, 264 Avenue St Pierre, 13005 Marseille, France; (A.F.); (F.C.); (J.-H.C.); (R.L.)
- Laboratory ADES, Aix-Marseille University, CNRS, EFS, 13005 Marseille, France;
| |
Collapse
|
12
|
Hao M, Xue L, Wen X, Sun L, Zhang L, Xing K, Hu X, Xu J, Xing D. Advancing bone regeneration: Unveiling the potential of 3D cell models in the evaluation of bone regenerative materials. Acta Biomater 2024; 183:1-29. [PMID: 38815683 DOI: 10.1016/j.actbio.2024.05.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/01/2024]
Abstract
Bone, a rigid yet regenerative tissue, has garnered extensive attention for its impressive healing abilities. Despite advancements in understanding bone repair and creating treatments for bone injuries, handling nonunions and large defects remains a major challenge in orthopedics. The rise of bone regenerative materials is transforming the approach to bone repair, offering innovative solutions for nonunions and significant defects, and thus reshaping orthopedic care. Evaluating these materials effectively is key to advancing bone tissue regeneration, especially in difficult healing scenarios, making it a critical research area. Traditional evaluation methods, including two-dimensional cell models and animal models, have limitations in predicting accurately. This has led to exploring alternative methods, like 3D cell models, which provide fresh perspectives for assessing bone materials' regenerative potential. This paper discusses various techniques for constructing 3D cell models, their pros and cons, and crucial factors to consider when using these models to evaluate bone regenerative materials. We also highlight the significance of 3D cell models in the in vitro assessments of these materials, discuss their current drawbacks and limitations, and suggest future research directions. STATEMENT OF SIGNIFICANCE: This work addresses the challenge of evaluating bone regenerative materials (BRMs) crucial for bone tissue engineering. It explores the emerging role of 3D cell models as superior alternatives to traditional methods for assessing these materials. By dissecting the construction, key factors of evaluating, advantages, limitations, and practical considerations of 3D cell models, the paper elucidates their significance in overcoming current evaluation method shortcomings. It highlights how these models offer a more physiologically relevant and ethically preferable platform for the precise assessment of BRMs. This contribution is particularly significant for "Acta Biomaterialia" readership, as it not only synthesizes current knowledge but also propels the discourse forward in the search for advanced solutions in bone tissue engineering and regeneration.
Collapse
Affiliation(s)
- Minglu Hao
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China.
| | - Linyuan Xue
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China
| | - Xiaobo Wen
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China
| | - Li Sun
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China
| | - Lei Zhang
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, Ontario N2L3G1, Canada
| | - Kunyue Xing
- Alliance Manchester Business School, The University of Manchester, Manchester M139PL, UK
| | - Xiaokun Hu
- Department of Interventional Medical Center, Affiliated Hospital of Qingdao University, Qingdao 26600, China
| | - Jiazhen Xu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China.
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
13
|
Mahmoudi N, Roque M, Paiva Dos Santos B, Oliveira H, Siadous R, Rey S, Garanger E, Lecommandoux S, Catros S, Garbay B, Amédée Vilamitjana J. An Elastin-Derived Composite Matrix for Enhanced Vascularized and Innervated Bone Tissue Reconstruction: From Material Development to Preclinical Evaluation. Adv Healthc Mater 2024; 13:e2303765. [PMID: 38651610 DOI: 10.1002/adhm.202303765] [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: 10/29/2023] [Revised: 03/28/2024] [Indexed: 04/25/2024]
Abstract
Despite progress in bone tissue engineering, reconstruction of large bone defects remains an important clinical challenge. Here, a biomaterial designed to recruit bone cells, endothelial cells, and neuronal fibers within the same matrix is developed, enabling bone tissue regeneration. The bioactive matrix is based on modified elastin-like polypeptides (ELPs) grafted with laminin-derived adhesion peptides IKVAV and YIGSR, and the SNA15 peptide for retention of hydroxyapatite (HA) particles. The composite matrix shows suitable porosity, interconnectivity, biocompatibility for endothelial cells, and the ability to support neurites outgrowth by sensory neurons. Subcutaneous implantation leads to the formation of osteoid tissue, characterized by the presence of bone cells, vascular networks, and neuronal structures, while minimizing inflammation. Using a rat femoral condyle defect model, longitudinal micro-CT analysis is performed, which demonstrates a significant increase in the volume of mineralized tissue when using the ELP-based matrix compared to empty defects and a commercially available control (Collapat). Furthermore, visible blood vessel networks and nerve fibers are observed within the lesions after a period of two weeks. By incorporating multiple key components that support cell growth, mineralization, and tissue integration, this ELP-based composite matrix provides a holistic and versatile solution to enhance bone tissue regeneration.
Collapse
Affiliation(s)
- Nadia Mahmoudi
- Tissue Bioengineering Laboratory (BioTis), Inserm U1026, University of Bordeaux, Bordeaux, France
| | - Micaela Roque
- Tissue Bioengineering Laboratory (BioTis), Inserm U1026, University of Bordeaux, Bordeaux, France
| | - Bruno Paiva Dos Santos
- Tissue Bioengineering Laboratory (BioTis), Inserm U1026, University of Bordeaux, Bordeaux, France
| | - Hugo Oliveira
- Tissue Bioengineering Laboratory (BioTis), Inserm U1026, University of Bordeaux, Bordeaux, France
| | - Robin Siadous
- Tissue Bioengineering Laboratory (BioTis), Inserm U1026, University of Bordeaux, Bordeaux, France
| | - Sylvie Rey
- Tissue Bioengineering Laboratory (BioTis), Inserm U1026, University of Bordeaux, Bordeaux, France
| | | | | | - Sylvain Catros
- CHU Bordeaux, Dentistry and Oral Health Department, Bordeaux, 33076, France
| | - Bertrand Garbay
- Univ. Bordeaux, CNRS, Bordeaux INP, LCPO, UMR, Pessac, 5629, France
| | | |
Collapse
|
14
|
Liu Z, Liu H, Liu S, Li B, Liu Y, Luo E. SIRT1 activation promotes bone repair by enhancing the coupling of type H vessel formation and osteogenesis. Cell Prolif 2024; 57:e13596. [PMID: 38211965 PMCID: PMC11150139 DOI: 10.1111/cpr.13596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/10/2023] [Accepted: 12/18/2023] [Indexed: 01/13/2024] Open
Abstract
Bone repair is intricately correlated with vascular regeneration, especially of type H vessels. Sirtuin 1 (SIRT1) expression is closely associated with endothelial function and vascular regeneration; however, the role of SIRT1 in enhancing the coupling of type H vessel formation with osteogenesis to promote bone repair needs to be investigated. A co-culture system combining human umbilical vein endothelial cells and osteoblasts was constructed, and a SIRT1 agonist was used to evaluate the effects of SIRT1 activity. The angiogenic and osteogenic capacities of the co-culture system were examined using short interfering RNA. Mouse models with bone defects in the femur or mandible were established to explore changes in type H vessel formation and bone repair following modulated SIRT1 activity. SIRT1 activation augmented the angiogenic and osteogenic capacities of the co-culture system by activating the PI3K/AKT/FOXO1 signalling pathway and did not significantly regulate osteoblast differentiation. Inhibition of the PI3K/AKT/FOXO1 pathway attenuated SIRT1-mediated effects. The SIRT1 activity in bone defects was positively correlated with the formation of type H vessels and bone repair in vivo, whereas SIRT1 inhibition substantially weakened vascular and bone formation. Thus, SIRT1 is crucial to the coupling of type H vessels with osteogenesis during bone repair.
Collapse
Affiliation(s)
- Zhikai Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Hanghang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Shibo Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Bolun Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yao Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - En Luo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
15
|
Xu J, He S, Xia T, Shan Y, Wang L. Targeting type H vessels in bone-related diseases. J Cell Mol Med 2024; 28:e18123. [PMID: 38353470 PMCID: PMC10865918 DOI: 10.1111/jcmm.18123] [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: 06/26/2023] [Revised: 01/01/2024] [Accepted: 01/09/2024] [Indexed: 02/16/2024] Open
Abstract
Blood vessels are essential for bone development and metabolism. Type H vessels in bone, named after their high expression of CD31 and Endomucin (Emcn), have recently been reported to locate mainly in the metaphysis, exhibit different molecular properties and couple osteogenesis and angiogenesis. A strong correlation between type H vessels and bone metabolism is now well-recognized. The crosstalk between type H vessels and osteoprogenitor cells is also involved in bone metabolism-related diseases such as osteoporosis, osteoarthritis, fracture healing and bone defects. Targeting the type H vessel formation may become a new approach for managing a variety of bone diseases. This review highlighted the roles of type H vessels in bone-related diseases and summarized the research attempts to develop targeted intervention, which will help us gain a better understanding of their potential value in clinical application.
Collapse
Affiliation(s)
- Juan Xu
- Outpatient DepartmentChildren's Hospital of Soochow UniversitySuzhouChina
| | - Shuang‐jian He
- Department of OrthopaedicsSuzhou Hospital, Affiliated Hospital of Medical School, Nanjing UniversitySuzhouChina
| | - Ting‐ting Xia
- Clinical Research InstituteSuzhou Hospital, Affiliated Hospital of Medical School, Nanjing UniversitySuzhouChina
| | - Yu Shan
- Department of OrthopeadicsSuzhou Ninth Hospital Affiliated to Soochow UniversitySuzhouChina
| | - Liang Wang
- Department of OrthopaedicsSuzhou Hospital, Affiliated Hospital of Medical School, Nanjing UniversitySuzhouChina
- Department of OrthopeadicsThe Fourth Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
16
|
Ji X, Bei HP, Zhong G, Shao H, He X, Qian X, Zhang Y, Zhao X. Premetastatic Niche Mimicking Bone-On-A-Chip: A Microfluidic Platform to Study Bone Metastasis in Cancer Patients. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2207606. [PMID: 37605335 DOI: 10.1002/smll.202207606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 07/08/2023] [Indexed: 08/23/2023]
Abstract
Primary cancer modulates the bone microenvironment to sow the seeds of dormancy and metastasis in tumor cells, leading to multiple organ metastasis and death. In this study, 3D printing and bone-on-a-chip (BOC) are combined to develop a BOC platform that mimics the pre-metastatic niches (PMNs) and facilitates elucidation of the interactions between bone-resident cells and metastatic tumor cells under the influence of primary cancer. Photocrosslinkable gelatin methacrylate (GelMA) is used as a 3D culturing hydrogel to encapsulate cells, and circulate tumor culture medium (CM) adjacent to the hydrogel to verify the critical role of mesenchymal stem cells (MSCs) and osteoclasts (RAW264.7s). Three niches: the dormancy niche, the perivascular niche, and the "vicious cycle" niche, are devised to recapitulate bone metastasis in one chip with high cell viability and excellent nutrient exchange. With respect to tumor dormancy and reactivation, the invadopodia formation of A549 lung cancer cells in communication with MSCs and RAW264.7 via the cortactin pathway is researched. As a proof of concept, the functionality and practicality of the platform are demonstrated by analyzing the invadopodia formation and the influence of various cells, and the establishment of the dynamic niches paves the way to understanding PMN formation and related drug discovery.
Collapse
Affiliation(s)
- Xiongfa Ji
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong SAR, China
- Department of Orthopaedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518057, China
| | - Ho-Pan Bei
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong SAR, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518057, China
| | - Guoqing Zhong
- Department of Orthopaedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China
| | - Hongwei Shao
- Department of Orthopaedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China
| | - Xuecheng He
- Department of Orthopaedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China
| | - Xin Qian
- Department of Orthopaedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China
| | - Yu Zhang
- Department of Orthopaedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080, Guangzhou, China
| | - Xin Zhao
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong SAR, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518057, China
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, 999077, Hong Kong SAR, China
| |
Collapse
|
17
|
Suo J, Gan Y, Xie Y, Xu S, Wang J, Chen D, Chen L, Deng L, Feng S, Han JJ, Jiang Q, Lei G, Liu P, Luo X, Ma X, Qu J, Song C, Tang P, Tang T, Wang S, Wei X, Wu C, Xiao G, Yang L, Zhang L, Zhang W, Zhang Z, Liu GH, Zhang C, Pei G, Luo J, Yue R, Zou W. A framework of biomarkers for skeletal aging: a consensus statement by the Aging Biomarker Consortium. LIFE MEDICINE 2023; 2:lnad045. [PMID: 39872060 PMCID: PMC11748998 DOI: 10.1093/lifemedi/lnad045] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/01/2023] [Indexed: 01/29/2025]
Abstract
The skeleton is an important structural and metabolic organ in human body, while aging is the physiological basis for degenerative skeletal diseases. China has the largest aging population in the world and faces great challenges in preventing and managing diseases related to skeletal aging. To address these challenges, the Aging China Biomarkers Consortium (ABC) has reached an expert consensus on biomarkers of skeletal aging by synthesizing the literature and insights from scientists and clinicians. The consensus provides a comprehensive assessment of biomarkers associated with skeletal aging and proposes a systematic framework that categorizes biomarkers into three dimensions, namely, functional, structural, and humoral dimensions. Within each dimension, the ABC recommended clinical and evidential research-based biomarkers for physiological aging and degenerative pathologies of the skeleton. This expert consensus aims to lay the foundation for future studies to assess the prediction, diagnosis, early warning, and treatment of diseases associated with skeletal aging, with the ultimate goal of improving the skeletal health of elderly populations in China and around the world.
Collapse
Affiliation(s)
- Aging Biomarker Consortium
- Department of Orthopedic Surgery, Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinlong Suo
- Department of Orthopedic Surgery, Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yibo Gan
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yangli Xie
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair (CBMR), Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shuqin Xu
- Department of Osteoporosis and Bone Diseases, Shanghai Clinical Research Center of Bone Disease, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianfang Wang
- Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Di Chen
- Research Center for Computer-Aided Drug Discovery, Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Lin Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair (CBMR), Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Lianfu Deng
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Department of Orthopaedics, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shiqing Feng
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jingdong Jackie Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing, China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Drum Tower Hospital affiliated to Medical School of Nanjing University, Nanjing, China
| | - Guanghua Lei
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Hunan Key Laboratory of Joint Degeneration and Injury, Department of Orthopaedics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Peng Liu
- State Key Laboratory of Trauma and Chemical Poisoning, Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xianghang Luo
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Department of Endocrinology, Endocrinology Research Center, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xin Ma
- Department of Orthopaedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Institute for Stem Cell and Regeneration, Institute for Stem Cell and Regenerative Medicine, University of Chinese Academy of Sciences, Beijing, China
| | - Chunli Song
- Beijing Key Laboratory of Spinal Disease, Department of Orthopedics, Engineering Research Center of Bone and Joint Precision Medicine, Peking University Third Hospital, Beijing, China
| | - Peifu Tang
- Department of Orthopaedic Trauma, the Fourth Medical Center, National Clinical Research Center for Orthopaedics & Sports Rehabilitation in China, Chinese PLA General Hospital, Beijing, China
| | - Tingting Tang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sijia Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiaochun Wei
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopedics, the Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
| | - Guozhi Xiao
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Liu Yang
- Institute of Orthopedic Surgery, Xijing Hospital, The Fourth Military Medical University, Xi’an, Shaanxi, China
- Medical Research Institute, Northwestern Polytechnical University, Xi’an, China
| | - Licheng Zhang
- Department of Orthopaedic Trauma, the Fourth Medical Center, National Clinical Research Center for Orthopaedics & Sports Rehabilitation in China, Chinese PLA General Hospital, Beijing, China
| | - Weiqi Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
| | - Zhenlin Zhang
- Department of Osteoporosis and Bone Diseases, Shanghai Clinical Research Center of Bone Disease, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Institute for Stem Cell and Regeneration, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Changqing Zhang
- Department of Orthopedic Surgery, Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gang Pei
- Collaborative Innovation Center for Brain Science, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Jian Luo
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, China
| | - Rui Yue
- Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Weiguo Zou
- Department of Orthopedic Surgery, Institute of Microsurgery on Extremities, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| |
Collapse
|
18
|
Dutta SD, Ganguly K, Patil TV, Randhawa A, Lim KT. Unraveling the potential of 3D bioprinted immunomodulatory materials for regulating macrophage polarization: State-of-the-art in bone and associated tissue regeneration. Bioact Mater 2023; 28:284-310. [PMID: 37303852 PMCID: PMC10248805 DOI: 10.1016/j.bioactmat.2023.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 04/29/2023] [Accepted: 05/20/2023] [Indexed: 06/13/2023] Open
Abstract
Macrophage-assisted immunomodulation is an alternative strategy in tissue engineering, wherein the interplay between pro-inflammatory and anti-inflammatory macrophage cells and body cells determines the fate of healing or inflammation. Although several reports have demonstrated that tissue regeneration depends on spatial and temporal regulation of the biophysical or biochemical microenvironment of the biomaterial, the underlying molecular mechanism behind immunomodulation is still under consideration for developing immunomodulatory scaffolds. Currently, most fabricated immunomodulatory platforms reported in the literature show regenerative capabilities of a particular tissue, for example, endogenous tissue (e.g., bone, muscle, heart, kidney, and lungs) or exogenous tissue (e.g., skin and eye). In this review, we briefly introduced the necessity of the 3D immunomodulatory scaffolds and nanomaterials, focusing on material properties and their interaction with macrophages for general readers. This review also provides a comprehensive summary of macrophage origin and taxonomy, their diverse functions, and various signal transduction pathways during biomaterial-macrophage interaction, which is particularly helpful for material scientists and clinicians for developing next-generation immunomodulatory scaffolds. From a clinical standpoint, we briefly discussed the role of 3D biomaterial scaffolds and/or nanomaterial composites for macrophage-assisted tissue engineering with a special focus on bone and associated tissues. Finally, a summary with expert opinion is presented to address the challenges and future necessity of 3D bioprinted immunomodulatory materials for tissue engineering.
Collapse
Affiliation(s)
- Sayan Deb Dutta
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Keya Ganguly
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Tejal V. Patil
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Aayushi Randhawa
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Ki-Taek Lim
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| |
Collapse
|
19
|
Wang SS, Zhu XX, Wu XY, Zhang WW, Ding YD, Jin SW, Zhang PH. Interaction Between Blood Vasculatures and Lymphatic Vasculatures During Inflammation. J Inflamm Res 2023; 16:3271-3281. [PMID: 37560514 PMCID: PMC10408656 DOI: 10.2147/jir.s414891] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 07/21/2023] [Indexed: 08/11/2023] Open
Abstract
Physiological activity cannot be regulated without the blood and lymphatic vasculatures, which play complementary roles in maintaining the body's homeostasis and immune responses. Inflammation is the body's initial response to pathological injury and is responsible for protecting the body, removing damaged tissues, and restoring and maintaining homeostasis in the body. A growing number of researches have shown that blood and lymphatic vessels play an essential role in a variety of inflammatory diseases. In the inflammatory state, the permeability of blood vessels and lymphatic vessels is altered, and angiogenesis and lymphangiogenesis subsequently occur. The blood vascular and lymphatic vascular systems interact to determine the development or resolution of inflammation. In this review, we discuss the changes that occur in the blood vascular and lymphatic vascular systems of several organs during inflammation, describe the different scenarios of angiogenesis and lymphangiogenesis at different sites of inflammation, and demonstrate the prospect of targeting the blood vasculature and lymphatic vasculature systems to limit the development of inflammation and promote the resolution of inflammation in inflammatory diseases.
Collapse
Affiliation(s)
- Shun-Shun Wang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
- Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
| | - Xin-Xu Zhu
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
- Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
| | - Xin-Yi Wu
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
- Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
| | - Wen-Wu Zhang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
- Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
| | - Yang-Dong Ding
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
- Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
| | - Sheng-Wei Jin
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
- Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
| | - Pu-Hong Zhang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
- Key Laboratory of Anesthesiology of Zhejiang Province, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
| |
Collapse
|
20
|
Watanabe H, Maishi N, Hoshi-Numahata M, Nishiura M, Nakanishi-Kimura A, Hida K, Iimura T. Skeletal-Vascular Interactions in Bone Development, Homeostasis, and Pathological Destruction. Int J Mol Sci 2023; 24:10912. [PMID: 37446097 DOI: 10.3390/ijms241310912] [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: 06/12/2023] [Revised: 06/28/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Bone is a highly vascularized organ that not only plays multiple roles in supporting the body and organs but also endows the microstructure, enabling distinct cell lineages to reciprocally interact. Recent studies have uncovered relevant roles of the bone vasculature in bone patterning, morphogenesis, homeostasis, and pathological bone destruction, including osteoporosis and tumor metastasis. This review provides an overview of current topics in the interactive molecular events between endothelial cells and bone cells during bone ontogeny and discusses the future direction of this research area to find novel ways to treat bone diseases.
Collapse
Affiliation(s)
- Haruhisa Watanabe
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Nako Maishi
- Department of Vascular Biology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Marie Hoshi-Numahata
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Mai Nishiura
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Atsuko Nakanishi-Kimura
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Kyoko Hida
- Department of Vascular Biology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Tadahiro Iimura
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| |
Collapse
|
21
|
Farahzadi R, Valipour B, Montazersaheb S, Fathi E. Targeting the stem cell niche micro-environment as therapeutic strategies in aging. Front Cell Dev Biol 2023; 11:1162136. [PMID: 37274742 PMCID: PMC10235764 DOI: 10.3389/fcell.2023.1162136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 05/02/2023] [Indexed: 06/06/2023] Open
Abstract
Adult stem cells (ASCs) reside throughout the body and support various tissue. Owing to their self-renewal capacity and differentiation potential, ASCs have the potential to be used in regenerative medicine. Their survival, quiescence, and activation are influenced by specific signals within their microenvironment or niche. In better words, the stem cell function is significantly influenced by various extrinsic signals derived from the niche. The stem cell niche is a complex and dynamic network surrounding stem cells that plays a crucial role in maintaining stemness. Studies on stem cell niche have suggested that aged niche contributes to the decline in stem cell function. Notably, functional loss of stem cells is highly associated with aging and age-related disorders. The stem cell niche is comprised of complex interactions between multiple cell types. Over the years, essential aspects of the stem cell niche have been revealed, including cell-cell contact, extracellular matrix interaction, soluble signaling factors, and biochemical and biophysical signals. Any alteration in the stem cell niche causes cell damage and affects the regenerative properties of the stem cells. A pristine stem cell niche might be essential for the proper functioning of stem cells and the maintenance of tissue homeostasis. In this regard, niche-targeted interventions may alleviate problems associated with aging in stem cell behavior. The purpose of this perspective is to discuss recent findings in the field of stem cell aging, heterogeneity of stem cell niches, and impact of age-related changes on stem cell behavior. We further focused on how the niche affects stem cells in homeostasis, aging, and the progression of malignant diseases. Finally, we detail the therapeutic strategies for tissue repair, with a particular emphasis on aging.
Collapse
Affiliation(s)
- Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behnaz Valipour
- Department of Anatomical Sciences, Sarab Faculty of Medical Sciences, Sarab, Iran
| | - Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| |
Collapse
|
22
|
Abstract
Despite advancement in therapeutic options, Non-Small Cell lung cancer (NSCLC) remains a lethal disease mostly due to late diagnosis at metastatic phase and drug resistance. Bone is one of the more frequent sites for NSCLC metastatization. A defined subset of cancer stem cells (CSCs) that possess motile properties, mesenchymal features and tumor initiation potential are defined as metastasis initiating cells (MICs). A better understanding of the mechanisms supporting MIC dissemination and interaction with bone microenvironment is fundamental to design novel rational therapeutic option for long lasting efficient treatment of NSCLC. In this review we will summarize findings about bone metastatic process initiated by NSCLC MICs. We will review how MICs can reach bone and interact with its microenvironment that supports their extravasation, seeding, dormancy/proliferation. The role of different cell types inside the bone metastatic niche, such as endothelial cells, bone cells, hematopoietic stem cells and immune cells will be discussed in regards of their impact in dictating the success of metastasis establishment by MICs. Finally, novel therapeutic options to target NSCLC MIC-induced bone metastases, increasing the survival of patients, will be presented.
Collapse
|
23
|
Interplay between fat cells and immune cells in bone: Impact on malignant progression and therapeutic response. Pharmacol Ther 2022; 238:108274. [DOI: 10.1016/j.pharmthera.2022.108274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 08/11/2022] [Accepted: 08/23/2022] [Indexed: 11/20/2022]
|
24
|
Zhang S, Tuk B, van de Peppel J, Kremers GJ, Koedam M, Pesch GR, Rahman Z, Hoogenboezem RM, Bindels EMJ, van Neck JW, Boukany PE, van Leeuwen JPTM, van der Eerden BCJ. Microfluidic evidence of synergistic effects between mesenchymal stromal cell-derived biochemical factors and biomechanical forces to control endothelial cell function. Acta Biomater 2022; 151:346-359. [PMID: 35995408 DOI: 10.1016/j.actbio.2022.08.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/20/2022] [Accepted: 08/12/2022] [Indexed: 11/01/2022]
Abstract
A functional vascular system is a prerequisite for bone repair as disturbed angiogenesis often causes non-union. Paracrine factors released from human bone marrow derived mesenchymal stromal cells (BMSCs) have angiogenic effects on endothelial cells. However, whether these paracrine factors participate in blood flow dynamics within bone capillaries remains poorly understood. Here, we used two different microfluidic designs to investigate critical steps during angiogenesis and found pronounced effects of endothelial cell proliferation as well as chemotactic and mechanotactic migration induced by BMSC conditioned medium (CM). The application of BMSC-CM in dynamic cultures demonstrates that bioactive factors in combination with fluidic flow-induced biomechanical signals significantly enhanced endothelial cell migration. Transcriptional analyses of endothelial cells demonstrate the induction of a unique gene expression profile related to tricarboxylic acid cycle and energy metabolism by the combination of BMSC-CM factors and shear stress, which opens an interesting avenue to explore during fracture healing. Our results stress the importance of in vivo - like microenvironments simultaneously including biochemical, biomechanical and oxygen levels when investigating key events during vessel repair. STATEMENT OF SIGNIFICANCE: Our results demonstrate the importance of recapitulating in vivo - like microenvironments when investigating key events during vessel repair. Endothelial cells exhibit enhanced angiogenesis characteristics when simultaneous exposing them to hMSC-CM, mechanical forces and biochemical signals simultaneously. The improved angiogenesis may not only result from the direct effect of growth factors, but also by reprogramming of endothelial cell metabolism. Moreover, with this model we demonstrated a synergistic impact of mechanical forces and biochemical factors on endothelial cell behavior and the expression of genes involved in the TCA cycle and energy metabolism, which opens an interesting new avenue to stimulate angiogenesis during fracture healing.
Collapse
Affiliation(s)
- Shuang Zhang
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Bastiaan Tuk
- Department of Plastic and Reconstructive Surgery, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Jeroen van de Peppel
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Gert-Jan Kremers
- Erasmus Optical Imaging Center, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Marijke Koedam
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Georg R Pesch
- Department of Chemical Engineering, Delft University of Technology; Delft, the Netherlands
| | - Zaid Rahman
- Department of Chemical Engineering, Delft University of Technology; Delft, the Netherlands
| | - Remco M Hoogenboezem
- Department of Hematology, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Eric M J Bindels
- Department of Hematology, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Johan W van Neck
- Department of Plastic and Reconstructive Surgery, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Pouyan E Boukany
- Department of Chemical Engineering, Delft University of Technology; Delft, the Netherlands
| | - Johannes P T M van Leeuwen
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center; Rotterdam, the Netherlands
| | - Bram C J van der Eerden
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center; Rotterdam, the Netherlands.
| |
Collapse
|
25
|
Di Maggio N, Banfi A. The osteo-angiogenic signaling crosstalk for bone regeneration: harmony out of complexity. Curr Opin Biotechnol 2022; 76:102750. [PMID: 35841865 DOI: 10.1016/j.copbio.2022.102750] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/05/2022] [Accepted: 05/31/2022] [Indexed: 11/03/2022]
Abstract
In recent years it has been increasingly appreciated that blood vessels are not simply suppliers of nutrients and oxygen, but actually play an exquisite regulatory role in bone development and repair. A specialized kind of endothelium, named type H because of its high expression of CD31 and Endomucin, constitutes anatomically defined vessels in proximity of the epiphyseal growth plate. Type H endothelium regulates the proliferation and differentiation of both osteoblasts and osteoclasts through the secretion of angiocrine signals and is a hub for the bidirectional molecular crosstalk between the different cell populations of the osteogenic microenvironment. Type H vessels are a key target for current translational approaches aiming at coupling angiogenesis and osteogenesis for bone repair. Open questions remain about their presence and features in notstereotyped tissues, like engineered osteogenic grafts, and the opportunities for their clinical stimulation by pharmacological treatments.
Collapse
Affiliation(s)
- Nunzia Di Maggio
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland.
| | - Andrea Banfi
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland; Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Switzerland.
| |
Collapse
|
26
|
Itkin T, Duarte D, Passaro D. Editorial: The Dynamic Interface Between Vascular Blood Vessels to Blood Forming Hematopoietic Stem Cells in Health and Disease. Front Cell Dev Biol 2022; 10:870129. [PMID: 35309923 PMCID: PMC8930838 DOI: 10.3389/fcell.2022.870129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 02/11/2022] [Indexed: 12/01/2022] Open
Affiliation(s)
- Tomer Itkin
- Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- Correspondence: Tomer Itkin, ; Delfim Duarte, ; Diana Passaro,
| | - Delfim Duarte
- Hematopoeisis and Microenvironments Group, Instituto de Investigação e Inovação Em Saúde (i3S), Universidade Do Porto, Porto, Portugal
- Department of Onco-Hematology, Instituto Português de Oncologia (IPO)-Porto, Porto, Portugal
- Department of Biomedicine, Unit of Biochemistry, Faculdade de Medicina da Universidade Do Porto, Porto, Portugal
- Correspondence: Tomer Itkin, ; Delfim Duarte, ; Diana Passaro,
| | - Diana Passaro
- Leukemia and Niche Dynamics Laboratory, Université de Paris, Institut Cochin, Institut National de La Santé et de La Recherche Médicale, Centre National de La Recherche Scientifique, Paris, France
- Correspondence: Tomer Itkin, ; Delfim Duarte, ; Diana Passaro,
| |
Collapse
|
27
|
Owen-Woods C, Kusumbe A. Fundamentals of bone vasculature: Specialization, interactions and functions. Semin Cell Dev Biol 2022; 123:36-47. [PMID: 34281770 DOI: 10.1016/j.semcdb.2021.06.025] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 02/06/2023]
Abstract
Angiogenesis, hematopoiesis and osteogenesis are fundamental processes mediating complex and essential biological functions. In the bone marrow, endothelial cells (ECs) are a principal mediator of regulatory signals that govern hematopoietic and mesenchymal stem cells. EC and osteoblast interactions and niche functions of ECs are fundamental in maintaining bone health and coordinating repair and regeneration following injury. These cellular interactions are subject to dysregulation and deterioration under stress, aging, chronic disease states and malignancy. Thus, the prospect of manipulating the bone vasculature has tremendous potential to advance therapeutic interventions for the management of bone diseases. This review discusses the current state of vascular-skeletal tissue interactions focusing on osteoblast and hematopoietic stem cells interaction with ECs.
Collapse
Affiliation(s)
- Charlotte Owen-Woods
- Tissue and Tumor Microenvironments Group, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| | - Anjali Kusumbe
- Tissue and Tumor Microenvironments Group, NDORMS, University of Oxford, Oxford OX3 7FY, UK.
| |
Collapse
|
28
|
Sekaran S, Thangavelu L. Re-appraising the role of flavonols, flavones and flavonones on osteoblasts and osteoclasts- A review on its molecular mode of action. Chem Biol Interact 2022; 355:109831. [PMID: 35120918 DOI: 10.1016/j.cbi.2022.109831] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 01/02/2022] [Accepted: 01/21/2022] [Indexed: 11/16/2022]
Abstract
Bone disorders have become a global concern illustrated with decreased bone mineral density and disruption in microarchitecture of natural bone tissue organization. Natural compounds that promote bone health by augmenting osteoblast functions and suppressing osteoclast functions has gained much attention and offer greater therapeutic value compared to conventional therapies. Amongst several plant-based molecules, flavonoids act as a major combatant in promoting bone health through their multi-faceted biological activities such as antioxidant, anti-inflammatory, and osteogenic properties. They protect bone loss by regulating the signalling cascades involved in osteoblast and osteoclast functions. Flavonoids augment osteoblastogenesis and inhibits osteoclastogenesis through their modulation of various signalling pathways. This review discusses the role of various flavonoids and their molecular mechanisms involved in maintaining bone health by regulating osteoblast and osteoclast functions.
Collapse
Affiliation(s)
- Saravanan Sekaran
- Centre for Trans-disciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute for Medical and Technical Sciences, Chennai, 600077, Tamil Nadu, India.
| | - Lakshmi Thangavelu
- Centre for Trans-disciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute for Medical and Technical Sciences, Chennai, 600077, Tamil Nadu, India
| |
Collapse
|
29
|
Jin X, Li Y, Yang Y, Shen H, Chen J, Xu B, Xu J. Thioacetamide promotes osteoclast transformation of bone marrow macrophages by influencing PI3K/AKT pathways. J Orthop Surg Res 2022; 17:53. [PMID: 35093114 PMCID: PMC8800259 DOI: 10.1186/s13018-022-02938-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/12/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Osteoclast cell increase is a major risk factor for osteoporosis and degenerative bone and joint diseases. At present, RANKL and M-CSF are commonly used to induce osteoclastogenesis. Thioacetamide (TAA) can lead to many types of liver and kidney damage, but less attention has been paid to the association of TAA with bone damage. In this work, we investigated the effects of TAA on the osteoclastogenesis and differentiation of bone marrow macrophages (BMMs).
Methods
BMMs of SD rat suckling mice were taken for primary culture. CCK-8 was used to detect the toxic effects of TAA on BMMs, and flow cytometry was used to detect the effects of TAA on the cell cycle, cell viability, apoptosis and intracytoplasmic Ca2+ concentration of BMMs. TRAP staining was used to detect the effect of RANKL and M-CSF and TAA on osteoclast differentiation of BMMs. Western Blot was used to detect the expression level of PI3K/AKT pathway and osteoclast-specific proteins (TRAP and cathepsin K).
Results
The results suggested that TAA inhibited the proliferation of BMMs, while enhancing osteoclastogenesis at 0.5 mg/mL and 1 mg/mL as assayed by TRAP staining. Exposed to TAA, BMMs could differentiate into osteoclast-like cells with overexpression of cathepsin K and TRAP proteins. Western blot results showed that TAA can activate the expression levels of P-PI3K, P-AKT, P-P38, and P-JNK, accompanied by apoptosis of BMMs and increase in intracellular Ca2+.
Conclusion
TAA may induce osteoclast formation in BMMs by activating the expression of PI3K/AKT pathway proteins, which is comparable to the classic osteoclast differentiation inducer RANKL and M-CSF. This suggests that we may find a cheap osteoclast inducer.
Collapse
|
30
|
Hematopoiesis, Inflammation and Aging-The Biological Background and Clinical Impact of Anemia and Increased C-Reactive Protein Levels on Elderly Individuals. J Clin Med 2022; 11:jcm11030706. [PMID: 35160156 PMCID: PMC8836692 DOI: 10.3390/jcm11030706] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 01/27/2023] Open
Abstract
Anemia and systemic signs of inflammation are common in elderly individuals and are associated with decreased survival. The common biological context for these two states is then the hallmarks of aging, i.e., genomic instability, telomere shortening, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion and altered intercellular communication. Such aging-associated alterations of hematopoietic stem cells are probably caused by complex mechanisms and depend on both the aging of hematopoietic (stem) cells and on the supporting stromal cells. The function of inflammatory or immunocompetent cells is also altered by aging. The intracellular signaling initiated by soluble proinflammatory mediators (e.g., IL1, IL6 and TNFα) is altered during aging and contributes to the development of both the inhibition of erythropoiesis with anemia as well as to the development of the acute-phase reaction as a systemic sign of inflammation with increased CRP levels. Both anemia and increased CRP levels are associated with decreased overall survival and increased cardiovascular mortality. The handling of elderly patients with inflammation and/or anemia should in our opinion be individualized; all of them should have a limited evaluation with regard to the cause of the abnormalities, but the extent of additional and especially invasive diagnostic evaluation should be based on an overall clinical evaluation and the possible therapeutic consequences.
Collapse
|
31
|
Wang Y, Li S, Zhao L, Cheng P, Liu J, Guo F, Xiao J, Zhu W, Chen A. Aging Relevant Metabolite Itaconate Inhibits Inflammatory Bone Loss. Front Endocrinol (Lausanne) 2022; 13:885879. [PMID: 35937818 PMCID: PMC9353012 DOI: 10.3389/fendo.2022.885879] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/07/2022] [Indexed: 11/13/2022] Open
Abstract
Progressive bone loss during aging makes osteoporosis one of the most common and life impacting conditions in geriatric populations. The bone homeostasis is maintained through persistent remodeling mediated by bone-forming osteoblast and bone-resorbing osteoclast. Inflammaging, a condition characterized by increased pro-inflammatory markers in the blood and other tissues during aging, has been reported to be associated with skeletal stem/progenitor cell dysfunction, which will result in impaired bone formation. However, the role of age-related inflammation and metabolites in regulation of osteoclast remains largely unknown. In the present study, we observed dichotomous phenotypes of anti-inflammatory metabolite itaconate in responding to inflammaging. Itaconate is upregulated in macrophages during aging but has less reactivity in responding to RANKL stimulation in aged macrophages. We confirmed the inhibitory effect of itaconate in regulating osteoclast differentiation and activation, and further verified the rescue role of itaconate in lipopolysaccharides induced inflammatory bone loss animal model. Our findings revealed that itaconate is a crucial regulatory metabolite during inflammaging that inhibits osteoclast to maintain bone homeostasis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Wentao Zhu
- *Correspondence: Wentao Zhu, ; Anmin Chen,
| | - Anmin Chen
- *Correspondence: Wentao Zhu, ; Anmin Chen,
| |
Collapse
|
32
|
Kumar N, Saraber P, Ding Z, Kusumbe AP. Diversity of Vascular Niches in Bones and Joints During Homeostasis, Ageing, and Diseases. Front Immunol 2021; 12:798211. [PMID: 34975909 PMCID: PMC8718446 DOI: 10.3389/fimmu.2021.798211] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/19/2021] [Indexed: 12/29/2022] Open
Abstract
The bones and joints in the skeletal system are composed of diverse cell types, including vascular niches, bone cells, connective tissue cells and mineral deposits and regulate whole-body homeostasis. The capacity of maintaining strength and generation of blood lineages lies within the skeletal system. Bone harbours blood and immune cells and their progenitors, and vascular cells provide several immune cell type niches. Blood vessels in bone are phenotypically and functionally diverse, with distinct capillary subtypes exhibiting striking changes with age. The bone vasculature has a special impact on osteogenesis and haematopoiesis, and dysregulation of the vasculature is associated with diverse blood and bone diseases. Ageing is associated with perturbed haematopoiesis, loss of osteogenesis, increased adipogenesis and diminished immune response and immune cell production. Endothelial and perivascular cells impact immune cell production and play a crucial role during inflammation. Here, we discuss normal and maladapted vascular niches in bone during development, homeostasis, ageing and bone diseases such as rheumatoid arthritis and osteoarthritis. Further, we discuss the role of vascular niches during bone malignancy.
Collapse
Affiliation(s)
| | | | | | - Anjali P. Kusumbe
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), Tissue and Tumor Microenvironments Group, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
33
|
Shang X, Böker KO, Taheri S, Lehmann W, Schilling AF. Extracellular Vesicles Allow Epigenetic Mechanotransduction between Chondrocytes and Osteoblasts. Int J Mol Sci 2021; 22:ijms222413282. [PMID: 34948080 PMCID: PMC8703680 DOI: 10.3390/ijms222413282] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/03/2021] [Accepted: 12/07/2021] [Indexed: 01/21/2023] Open
Abstract
MicroRNAs (miRNAs) can be transported in extracellular vesicles (EVs) and are qualified as possible messengers for cell–cell communication. In the context of osteoarthritis (OA), miR-221-3p has been shown to have a mechanosensitive and a paracrine function inside cartilage. However, the question remains if EVs with miR-221-3p can act as molecular mechanotransducers between cells of different tissues. Here, we studied the effect of EV-mediated transport in the communication between chondrocytes and osteoblasts in vitro in a rat model. In silico analysis (Targetscan, miRWalk, miRDB) revealed putative targets of miRNA-221-3p (CDKN1B/p27, TIMP-3, Tcf7l2/TCF4, ARNT). Indeed, transfection of miRNA-221-3p in chondrocytes and osteoblasts resulted in regulation of these targets. Coculture experiments of transfected chondrocytes with untransfected osteoblasts not only showed regulation of these target genes in osteoblasts but also inhibition of their bone formation capacity. Direct treatment with chondrocyte-derived EVs validated that chondrocyte-produced extracellular miR-221-3p was responsible for this effect. Altogether, our study provides a novel perspective on a possible communication pathway of a mechanically induced epigenetic signal through EVs. This may be important for processes at the interface of bone and cartilage, such as OA development, physiologic joint homeostasis, growth or fracture healing, as well as for other tissue interfaces with differing biomechanical properties.
Collapse
|
34
|
Zhou J, Zhang Z, Joseph J, Zhang X, Ferdows BE, Patel DN, Chen W, Banfi G, Molinaro R, Cosco D, Kong N, Joshi N, Farokhzad OC, Corbo C, Tao W. Biomaterials and nanomedicine for bone regeneration: Progress and future prospects. EXPLORATION (BEIJING, CHINA) 2021; 1:20210011. [PMID: 37323213 PMCID: PMC10190996 DOI: 10.1002/exp.20210011] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 08/12/2021] [Indexed: 06/14/2023]
Abstract
Bone defects pose a heavy burden on patients, orthopedic surgeons, and public health resources. Various pathological conditions cause bone defects including trauma, tumors, inflammation, osteoporosis, and so forth. Auto- and allograft transplantation have been developed as the most commonly used clinic treatment methods, among which autologous bone grafts are the golden standard. Yet the repair of bone defects, especially large-volume defects in the geriatric population or those complicated with systemic disease, is still a challenge for regenerative medicine from the clinical perspective. The fast development of biomaterials and nanomedicine favors the emergence and promotion of efficient bone regeneration therapies. In this review, we briefly summarize the progress of novel biomaterial and nanomedical approaches to bone regeneration and then discuss the current challenges that still hinder their clinical applications in treating bone defects.
Collapse
Affiliation(s)
- Jun Zhou
- Center for Nanomedicine and Department of AnesthesiologyBrigham and Women's Hospital Harvard Medical SchoolBostonMassachusettsUSA
| | - Zhongyang Zhang
- Center for Nanomedicine and Department of AnesthesiologyBrigham and Women's Hospital Harvard Medical SchoolBostonMassachusettsUSA
| | - John Joseph
- Center for Nanomedicine and Department of AnesthesiologyBrigham and Women's Hospital Harvard Medical SchoolBostonMassachusettsUSA
| | - Xingcai Zhang
- School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- School of EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Bijan Emiliano Ferdows
- Center for Nanomedicine and Department of AnesthesiologyBrigham and Women's Hospital Harvard Medical SchoolBostonMassachusettsUSA
- Pomona CollegeClaremontCaliforniaUSA
| | - Dylan Neal Patel
- Center for Nanomedicine and Department of AnesthesiologyBrigham and Women's Hospital Harvard Medical SchoolBostonMassachusettsUSA
- Jericho High SchoolJerichoNew YorkUSA
| | - Wei Chen
- Center for Nanomedicine and Department of AnesthesiologyBrigham and Women's Hospital Harvard Medical SchoolBostonMassachusettsUSA
| | - Giuseppe Banfi
- IRCCS GaleazziMilanoItaly
- Università Vita e Salute San RaffaeleMilanoItaly
| | | | - Donato Cosco
- Department of Health ScienceCampus Universitario‐Germaneto“Magna Græcia” University of CatanzaroCatanzaroItaly
| | - Na Kong
- Center for Nanomedicine and Department of AnesthesiologyBrigham and Women's Hospital Harvard Medical SchoolBostonMassachusettsUSA
| | - Nitin Joshi
- Center for Nanomedicine and Department of AnesthesiologyBrigham and Women's Hospital Harvard Medical SchoolBostonMassachusettsUSA
| | - Omid C. Farokhzad
- Center for Nanomedicine and Department of AnesthesiologyBrigham and Women's Hospital Harvard Medical SchoolBostonMassachusettsUSA
| | - Claudia Corbo
- School of Medicine and SurgeryNanomedicine Center NanomibUniversity of Milano‐BicoccaVedano al LambroItaly
| | - Wei Tao
- Center for Nanomedicine and Department of AnesthesiologyBrigham and Women's Hospital Harvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
35
|
The endothelium-bone axis in development, homeostasis and bone and joint disease. Nat Rev Rheumatol 2021; 17:608-620. [PMID: 34480164 DOI: 10.1038/s41584-021-00682-3] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2021] [Indexed: 01/20/2023]
Abstract
Blood vessels form a versatile transport network that is best known for its critical roles in processes such as tissue oxygenation, metabolism and immune surveillance. The vasculature also provides local, often organ-specific, molecular signals that control the behaviour of other cell types in their vicinity during development, homeostasis and regeneration, and also in disease processes. In the skeletal system, the local vasculature is actively involved in both bone formation and resorption. In addition, blood vessels participate in inflammatory processes and contribute to the pathogenesis of diseases that affect the joints, such as rheumatoid arthritis and osteoarthritis. This Review summarizes the current understanding of the architecture, angiogenic growth and functional properties of the bone vasculature. The effects of ageing and pathological conditions, including arthritis and osteoporosis, are also discussed.
Collapse
|
36
|
Belyavsky A, Petinati N, Drize N. Hematopoiesis during Ontogenesis, Adult Life, and Aging. Int J Mol Sci 2021; 22:ijms22179231. [PMID: 34502137 PMCID: PMC8430730 DOI: 10.3390/ijms22179231] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/13/2021] [Accepted: 08/24/2021] [Indexed: 12/17/2022] Open
Abstract
In the bone marrow of vertebrates, two types of stem cells coexist-hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs). Hematopoiesis only occurs when these two stem cell types and their descendants interact. The descendants of HSCs supply the body with all the mature blood cells, while MSCs give rise to stromal cells that form a niche for HSCs and regulate the process of hematopoiesis. The studies of hematopoiesis were initially based on morphological observations, later extended by the use of physiological methods, and were subsequently augmented by massive application of sophisticated molecular techniques. The combination of these methods produced a wealth of new data on the organization and functional features of hematopoiesis in the ontogenesis of mammals and humans. This review summarizes the current views on hematopoiesis in mice and humans, discusses the development of blood elements and hematopoiesis in the embryo, and describes how the hematopoietic system works in the adult organism and how it changes during aging.
Collapse
Affiliation(s)
- Alexander Belyavsky
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia;
| | | | - Nina Drize
- National Research Center for Hematology, 125167 Moscow, Russia;
- Correspondence:
| |
Collapse
|
37
|
Marrow failure and aging: The role of "Inflammaging". Best Pract Res Clin Haematol 2021; 34:101283. [PMID: 34404535 DOI: 10.1016/j.beha.2021.101283] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 07/02/2021] [Indexed: 02/07/2023]
Abstract
Despite aging and the enormous cellular output required of the marrow every day of the lifespan, most aged patients do not suffer significant marrow failure or cytopenias, an attestation to the proliferative capacity of this system. However, as marrow and its hematopoietic stem cells age, a reduction in ability to maintain homeostasis after stress or with exposure to prolonged chronic inflammation, so-called "inflammaging," may contribute to cytopenias, inadequate immune responses, and dysplasia/leukemia. In some instances, these changes may be intrinsic to the stem cell but in others, there may be extrinsic environmental influences. In this review, the role of aging as it relates to stem cell changes, immune function, and anemia are reviewed.
Collapse
|
38
|
Hofbauer LC, Bozec A, Rauner M, Jakob F, Perner S, Pantel K. Novel approaches to target the microenvironment of bone metastasis. Nat Rev Clin Oncol 2021; 18:488-505. [PMID: 33875860 DOI: 10.1038/s41571-021-00499-9] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2021] [Indexed: 02/07/2023]
Abstract
Bone metastases are a frequent and severe complication of advanced-stage cancers. Breast and prostate cancers, the most common malignancies in women and men, respectively, have a particularly high propensity to metastasize to bone. Conceptually, circulating tumour cells (CTCs) in the bloodstream and disseminated tumour cells (DTCs) in the bone marrow provide a snapshot of the dissemination and colonization process en route to clinically apparent bone metastases. Many cell types that constitute the bone microenvironment, including osteoblasts, osteocytes, osteoclasts, adipocytes, endothelial cells, haematopoietic stem cells and immune cells, engage in a dialogue with tumour cells. Some of these cells modify tumour biology, while others are disrupted and out-competed by tumour cells, thus leading to distinct phases of tumour cell migration, dormancy and latency, and therapy resistance and progression to overt bone metastases. Several current bone-protective therapies act by interrupting these interactions, mainly by targeting tumour cell-osteoclast interactions. In this Review, we describe the functional roles of the bone microenvironment and its components in the initiation and propagation of skeletal metastases, outline the biology and clinical relevance of CTCs and DTCs, and discuss established and future therapeutic approaches that specifically target defined components of the bone microenvironment to prevent or treat skeletal metastases.
Collapse
Affiliation(s)
- Lorenz C Hofbauer
- University Center for Healthy Aging, Dresden University of Technology, Dresden, Germany. .,Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany. .,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) partner site Dresden, Dresden, Germany.
| | - Aline Bozec
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg and University Hospital Erlangen, Erlangen, Germany
| | - Martina Rauner
- University Center for Healthy Aging, Dresden University of Technology, Dresden, Germany.,Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany
| | - Franz Jakob
- Department of Orthopedic Surgery, Julius Maximilians University of Würzburg, Würzburg, Germany.,Department of Functional Materials in Medicine and Dentistry, Julius Maximilians University of Würzburg, Würzburg, Germany
| | - Sven Perner
- Institute of Pathology, University Hospital of Schleswig-Holstein, Campus Lübeck, Lübeck, Germany.,Pathology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Klaus Pantel
- Department of Tumor Biology, Center of Experimental Medicine, University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
39
|
Wang F, Wang W, Kong L, Shi L, Wang M, Chai Y, Xu J, Kang Q. Accelerated Bone Regeneration by Adrenomedullin 2 Through Improving the Coupling of Osteogenesis and Angiogenesis via β-Catenin Signaling. Front Cell Dev Biol 2021; 9:649277. [PMID: 33937244 PMCID: PMC8079771 DOI: 10.3389/fcell.2021.649277] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/25/2021] [Indexed: 11/13/2022] Open
Abstract
Both osteogenic differentiation and the pro-angiogenic potential of bone marrow mesenchymal stem cells (BMSCs) contribute to bone regeneration during distraction osteogenesis (DO). Adrenomedullin 2 (ADM2), an endogenous bioactive peptide belonging to the calcitonin gene-related peptide family, exhibits various biological activities associated with the inhibition of inflammation and the attenuation of ischemic-hypoxic injury. However, the effects and underlying mechanisms of ADM2 in osteogenic differentiation and the pro-angiogenic potential of BMSCs, along with bone regeneration, remain poorly understood. In the present study, we found that osteogenic induction enhanced the pro-angiogenic potential of BMSCs, and ADM2 treatment further improved the osteogenic differentiation and pro-angiogenic potential of BMSCs. Moreover, the accumulation and activation of β-catenin, which is mediated by the inhibition of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and the activation of protein kinase B (AKT), have been shown to contribute to the effects of ADM2 on BMSCs. In vivo, ADM2 accelerated vessel expansion and bone regeneration, as revealed by improved radiological and histological manifestations and the biomechanical parameters in a rat DO model. Based on the present results, we concluded that ADM2 accelerates bone regeneration during DO by enhancing the osteogenic differentiation and pro-angiogenic potential of BMSCs, partly through the NF-κB/β-catenin and AKT/β-catenin pathways. Moreover, these findings imply that BMSC-mediated coupling of osteogenesis and angiogenesis may be a promising therapeutic strategy for DO patients.
Collapse
|
40
|
Stucker S, De Angelis J, Kusumbe AP. Heterogeneity and Dynamics of Vasculature in the Endocrine System During Aging and Disease. Front Physiol 2021; 12:624928. [PMID: 33767633 PMCID: PMC7987104 DOI: 10.3389/fphys.2021.624928] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 02/09/2021] [Indexed: 12/11/2022] Open
Abstract
The endocrine system consists of several highly vascularized glands that produce and secrete hormones to maintain body homeostasis and regulate a range of bodily functions and processes, including growth, metabolism and development. The dense and highly vascularized capillary network functions as the main transport system for hormones and regulatory factors to enable efficient endocrine function. The specialized capillary types provide the microenvironments to support stem and progenitor cells, by regulating their survival, maintenance and differentiation. Moreover, the vasculature interacts with endocrine cells supporting their endocrine function. However, the structure and niche function of vasculature in endocrine tissues remain poorly understood. Aging and endocrine disorders are associated with vascular perturbations. Understanding the cellular and molecular cues driving the disease, and age-related vascular perturbations hold potential to manage or even treat endocrine disorders and comorbidities associated with aging. This review aims to describe the structure and niche functions of the vasculature in various endocrine glands and define the vascular changes in aging and endocrine disorders.
Collapse
Affiliation(s)
| | | | - Anjali P. Kusumbe
- Tissue and Tumor Microenvironments Group, Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, United Kingdom
| |
Collapse
|