1
|
Wang Y, Zhang X, Li X, Cheng M, Cui X. The vascular microenvironment and its stem cells regulate vascular homeostasis. Front Cell Dev Biol 2025; 13:1544129. [PMID: 40114970 PMCID: PMC11922910 DOI: 10.3389/fcell.2025.1544129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
The vascular microenvironment comprises of anatomical structures, extracellular matrix components, and various cell populations, which play a crucial role in regulating vascular homeostasis and influencing vascular structure and function. Under physiological conditions, intrinsic regulation of the vascular microenvironment is required to sustain vascular homeostasis. In contrast, under pathological conditions, alterations to this microenvironment lead to vascular injury and pathological remodeling. According to the anatomy, the vascular microenvironment can be subdivided into three sections from the inside out. The vascular endothelial microenvironment, centered on vascular endothelial cells (VECs), includes the extracellular matrix and various vascular physicochemical factors. The VECs interact with vascular physicochemical factors to regulate the function of various parenchymal cells, including hepatocytes, neurons and tumor cells. The vascular wall microenvironment, comprising the vasa vasorum and their unique stem/progenitor cell niches, plays a pivotal role in vascular inflammation and pathological remodeling. Additionally, the perivascular microenvironment, which includes perivascular adipose tissue, consists of adipocytes and stem cells, which contribute to the pathological processes of atherosclerosis. It is anticipated that targeted regulation of the vascular microenvironment will emerge as a novel approach for the treatment of various diseases. Accordingly, this review will examine the structure of the vascular microenvironment, the regulation of vascular function by vascular cells and stem/progenitor cells, and the role of the vascular microenvironment in regulating cardiovascular diseases.
Collapse
Affiliation(s)
- Yanhui Wang
- Medical Physiology Laboratory, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Xiaoyun Zhang
- Medical Physiology Laboratory, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Xin Li
- Medical Physiology Laboratory, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Min Cheng
- Medical Physiology Laboratory, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Xiaodong Cui
- Medical Physiology Laboratory, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| |
Collapse
|
2
|
Melis S, Trompet D, Chagin AS, Maes C. Skeletal stem and progenitor cells in bone physiology, ageing and disease. Nat Rev Endocrinol 2025; 21:135-153. [PMID: 39379711 DOI: 10.1038/s41574-024-01039-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/30/2024] [Indexed: 10/10/2024]
Abstract
Skeletal stem cells (SSCs) and related progenitors with osteogenic potential, collectively termed skeletal stem and/or progenitor cells (SSPCs), are crucial for providing osteoblasts for bone formation during homeostatic tissue turnover and fracture repair. Besides mediating normal bone physiology, they also have important roles in various metabolic bone diseases, including osteoporosis. SSPCs are of tremendous interest because they represent prime future targets for osteoanabolic therapies and bone regenerative medicine. Remarkable progress has been made in characterizing various SSC and SSPC populations in postnatal bone. SSPCs exist in the periosteum and within the bone marrow stroma, including subsets localizing around arteriolar and sinusoidal blood vessels; they can display osteogenic, chondrogenic, adipogenic and/or fibroblastic potential, and exert critical haematopoiesis-supportive functions. However, much remains to be clarified. By the current markers, bona fide SSCs are commonly contained within broader SSPC populations characterized by considerable heterogeneity and overlap, whose common versus specific functions in health and disease have not been fully unravelled. Here, we review the present knowledge of the identity, fates and relationships of SSPC populations in the postnatal bone environment, their contributions to bone maintenance, the changes observed upon ageing, and the effect of metabolic diseases such as osteoporosis and diabetes mellitus.
Collapse
Affiliation(s)
- Seppe Melis
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Dana Trompet
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Andrei S Chagin
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Christa Maes
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium.
| |
Collapse
|
3
|
Yusoff NA, Abd Hamid Z, Budin SB, Taib IS. Hematopoietic stem cell discovery: unveiling the historical and future perspective of colony-forming units assay. PeerJ 2025; 13:e18854. [PMID: 39897489 PMCID: PMC11786707 DOI: 10.7717/peerj.18854] [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: 05/02/2024] [Accepted: 12/20/2024] [Indexed: 02/04/2025] Open
Abstract
Stem cells are special cells with the distinctive capability to self-renew, forming a new pool of undifferentiated stem cells. They are also able to differentiate into lineage-specific cell types that are specialized and matured. Thus, stem cells are considered as the building blocks of tissues and organs in which they reside. Among the many types of stem cells, hematopoietic stem cells (HSCs) are the most studied adult stem cells and are considered as a promising source of cells for applications in the clinical and basic sciences. Historically, research on HSCs was initiated in the 1940s, where in a groundbreaking experiment, intravenously injected bone marrow (BM) cells prevented the death of irradiated mice by restoring blood cell production. Since then, HSCs have been studied and utilized in medical therapies and research for over several decades. Over time, more sophisticated tools have been developed to evaluate the behaviour of specifically purified subsets of hematopoietic cells that have the capacity to produce blood cells. One of the established tools is the colony-forming units (CFUs) assay. This assay facilitates the identification, enumeration, and analysis of colonies formed by differentiated hematopoietic stem and progenitor cells (HSPCs) from myeloid, erythroid and lymphoid lineages. Hence, the CFUs assay is a fundamental in vitro platform that allows functional studies on the lineage potential of an individual HSPCs. The outcomes of such studies are crucial in providing critical insights into hematopoiesis. In this review, we explore the fundamental discoveries concerning the CFUs assay by covering the following aspects: (i) the historical overview of the CFUs assay for the study of clonal hematopoiesis involving multilineage potential of HSPCs, (ii) its use in various experimental models comprising humans, mice/rodents, zebrafish and induced pluripotent stem cells (iPSCs) and (iii) research gaps and future direction concerning the role of CFUs assay in clinical and basic sciences. Overall, the CFUs assay confers a transformative platform for a better understanding of HSPCs biology in governing hematopoiesis.
Collapse
Affiliation(s)
- Nur Afizah Yusoff
- Biomedical Science Programme and Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Zariyantey Abd Hamid
- Biomedical Science Programme and Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Siti Balkis Budin
- Biomedical Science Programme and Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Izatus Shima Taib
- Biomedical Science Programme and Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
4
|
Zheng Y, Cong L, Zhao L, Wang P, Xing L, Liu J, Xu H, Li N, Zhao Y, Yuan L, Shi Q, Sun X, Liang Q, Wang Y. Lymphatic platelet thrombosis limits bone repair by precluding lymphatic transporting DAMPs. Nat Commun 2025; 16:829. [PMID: 39827193 PMCID: PMC11742876 DOI: 10.1038/s41467-025-56147-8] [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: 10/21/2023] [Accepted: 01/05/2025] [Indexed: 01/22/2025] Open
Abstract
In the musculoskeletal system, lymphatic vessels (LVs), which are interdigitated with blood vessels, travel and form an extensive transport network. Blood vessels in bone regulate osteogenesis and hematopoiesis, however, whether LVs in bone affect fracture healing is unclear. Here, we investigate the lymphatic draining function at the tibial fracture sites using near-infrared indocyanine green lymphatic imaging (NIR-ICG) and discover that lymphatic drainage insufficiency (LDI) starts on day one and persists for up to two weeks following the fracture in male mice. Sufficient lymphatic drainage facilitates fracture healing in male mice. Furthermore, we identify that lymphatic platelet thrombosis (LPT) blocks the draining lymphoid sinus and LVs, causes LDI, and inhibits fracture healing in male mice, which can be rescued by a blood thinner. Moreover, unblocked lymphatic drainage decreases neutrophils and increases M2-type macrophages of the hematoma niche to support osteoblast (OB) survival and bone marrow-derived mesenchymal stem cell (BMSC) proliferation via transporting damage-associated molecular patterns (DAMPs) in male rats. Lymphatic platelet thrombolysis also benefits senile fracture healing in female mice. These findings demonstrate that LPT limits bone regeneration by impeding lymphatic transporting DAMPs. Together, these findings represent a way forward in the treatment of bone repair.
Collapse
Affiliation(s)
- Yangkang Zheng
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, 201203, Shanghai, China
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 227 Chongqing South Road, 200025, Shanghai, China
| | - Lin Cong
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, 201203, Shanghai, China
| | - Li Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, 201203, Shanghai, China
| | - Pengyu Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, 201203, Shanghai, China
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine and Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 227 Chongqing South Road, 200025, Shanghai, China
| | - Hao Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, 201203, Shanghai, China
| | - Ning Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, 201203, Shanghai, China
| | - Yongjian Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, 201203, Shanghai, China
| | - Luying Yuan
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, 201203, Shanghai, China
| | - Qi Shi
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, 201203, Shanghai, China
| | - Xueqing Sun
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 227 Chongqing South Road, 200025, Shanghai, China
| | - Qianqian Liang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China.
- Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China.
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, 201203, Shanghai, China.
| | - YongJun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China.
- Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, 200032, Shanghai, China.
- Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), 1200 Cailun Road, 201203, Shanghai, China.
| |
Collapse
|
5
|
Zhai Y, Zhou Z, Xing X, Nuzzle M, Zhang X. Differential bone and vessel type formation at superior and dura periosteum during cranial bone defect repair. Bone Res 2025; 13:8. [PMID: 39805832 PMCID: PMC11729862 DOI: 10.1038/s41413-024-00379-9] [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: 04/17/2024] [Revised: 08/14/2024] [Accepted: 10/09/2024] [Indexed: 01/16/2025] Open
Abstract
The cranial mesenchyme, originating from both neural crest and mesoderm, imparts remarkable regional specificity and complexity to postnatal calvarial tissue. While the distinct embryonic origins of the superior and dura periosteum of the cranial parietal bone have been described, the extent of their respective contributions to bone and vessel formation during adult bone defect repair remains superficially explored. Utilizing transgenic mouse models in conjunction with high-resolution multiphoton laser scanning microscopy (MPLSM), we have separately evaluated bone and vessel formation in the superior and dura periosteum before and after injury, as well as following intermittent treatment of recombinant peptide of human parathyroid hormone (rhPTH), Teriparatide. Our results show that new bone formation along the dura surface is three times greater than that along the superior periosteal surface following injury, regardless of Teriparatide treatment. Targeted deletion of PTH receptor PTH1R via SMA-CreER and Col 1a (2.3)-CreER results in selective reduction of bone formation, suggesting different progenitor cell pools in the adult superior and dura periosteum. Consistently, analyses of microvasculature show higher vessel density and better organized arterial-venous vessel network associated with a 10-fold more osteoblast clusters at dura periosteum as compared to superior periosteum. Intermittent rhPTH treatment further enhances the arterial vessel ratio at dura periosteum and type H vessel formation in cortical bone marrow space. Taken together, our study demonstrates a site-dependent coordinated osteogenic and angiogenic response, which is determined by regional osteogenic progenitor pool as well as the coupling blood vessel network at the site of cranial defect repair.
Collapse
Affiliation(s)
- Yuankun Zhai
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | - Zhuang Zhou
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | - Xiaojie Xing
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | - Mark Nuzzle
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | - Xinping Zhang
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA.
| |
Collapse
|
6
|
Boueya IL, Sandhow L, Albuquerque JRP, Znaidi R, Passaro D. Endothelial heterogeneity in bone marrow: insights across development, adult life and leukemia. Leukemia 2025; 39:8-24. [PMID: 39528790 PMCID: PMC11717709 DOI: 10.1038/s41375-024-02453-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: 07/24/2024] [Revised: 10/04/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
The central role of the endothelial microenvironment in orchestrating bone marrow (BM) homeostasis and hematopoietic support has been confirmed at various developmental stages and in adult life. The BM vasculature is crucial in mediating communication between BM parenchyma and circulating blood, displaying remarkable heterogeneity in structure and function. While vascular cell diversity in other tissues has long been recognized, the molecular basis of this phenomenon in BM is just now emerging. Over the past decade, single-cell approaches and microscopic observations have expanded our understanding of BM vasculature. While solely characterized for their paracrine properties in the past, recent advances have revolutionized our perception of endothelial function, revealing distinct anatomical locations associated with diverse endothelial cell states. The identification of phenotypic differences between normal and pathological conditions has therefore deepened our understanding of vascular dynamics and their impact on hematopoiesis in health and disease. In this review, we highlight key milestones and recent advances in understanding vascular heterogeneity within BM microenvironment during development, adulthood and aging. We also explore how leukemia affects this heterogeneity and how we can take this knowledge forward to improve clinical practices. By synthesizing existing literature, we aim to address unresolved questions and outline future research directions.
Collapse
Affiliation(s)
- I L Boueya
- Leukemia and Niche Dynamics laboratory, Institut Cochin, Université Paris Cité UMR-S1016, INSERM U1016, CNRS UMR8104, Paris, France
| | - L Sandhow
- Leukemia and Niche Dynamics laboratory, Institut Cochin, Université Paris Cité UMR-S1016, INSERM U1016, CNRS UMR8104, Paris, France
| | - J R P Albuquerque
- Leukemia and Niche Dynamics laboratory, Institut Cochin, Université Paris Cité UMR-S1016, INSERM U1016, CNRS UMR8104, Paris, France
| | - R Znaidi
- Leukemia and Niche Dynamics laboratory, Institut Cochin, Université Paris Cité UMR-S1016, INSERM U1016, CNRS UMR8104, Paris, France
| | - D Passaro
- Leukemia and Niche Dynamics laboratory, Institut Cochin, Université Paris Cité UMR-S1016, INSERM U1016, CNRS UMR8104, Paris, France.
| |
Collapse
|
7
|
Zhao K, Zhu GZ, Li HZ, Gao JW, Tu C, Wu DZ, Huang YS, Han D, Chen XY, Wu LY, Zhong ZM. Accumulation of Advanced Oxidation Protein Products Promotes Age-Related Decline of Type H Vessels in Bone. J Gerontol A Biol Sci Med Sci 2024; 80:glae271. [PMID: 39506899 DOI: 10.1093/gerona/glae271] [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: 01/06/2024] [Indexed: 11/08/2024] Open
Abstract
Type H vessels have been proven to couple angiogenesis and osteogenesis. The decline of type H vessels contributes to bone loss in the aging process. Aging is accompanied by the accumulation of advanced oxidation protein products (AOPPs). However, whether AOPP accumulation is involved in age-related decline of type H vessels is unclear. Here, we show that the increase of AOPP levels in plasma and bone was correlated with the decline of type H vessels and loss of bone mass in old mice. Exposure of microvascular endothelial cells to AOPPs significantly inhibited cell proliferation, migration, and tube formation; increased NADPH oxidase activity and excessive reactive oxygen species generation; upregulated the expression of vascular cell adhesion molecule-1 and intercellular cell adhesion molecule-1; and eventually impaired angiogenesis, which was alleviated by redox modulator N-acetylcysteine and NADPH oxidase inhibitor apocynin. Furthermore, reduced AOPP accumulation by NAC treatment was able to alleviate significantly the decline of type H vessels, bone mass loss, and deterioration of bone microstructure in old mice. Collectively, these findings suggest that AOPPs accumulation contributes to the decline of type H vessels in the aging process, and illuminate a novel potential mechanism underlying age-related bone loss.
Collapse
Affiliation(s)
- Kai Zhao
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
- Department of Orthopaedics, First Affiliated Hospital of Gannan Medical University, Ganzhou, People's Republic of China
| | - Guo-Zheng Zhu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Hong-Zhou Li
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Jia-Wen Gao
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Chen Tu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Di-Zheng Wu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Yu-Sheng Huang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Dong Han
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Xing-Yu Chen
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Long-Yan Wu
- Department of Ultrasound Medicine, Ganzhou People's Hospital, Ganzhou, People's Republic of China
| | - Zhao-Ming Zhong
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
8
|
Chow SKH, Gao Q, Pius A, Morita M, Ergul Y, Murayama M, Shinohara I, Cekuc MS, Ma C, Susuki Y, Goodman SB. The Advantages and Shortcomings of Stem Cell Therapy for Enhanced Bone Healing. Tissue Eng Part C Methods 2024; 30:415-430. [PMID: 39311464 DOI: 10.1089/ten.tec.2024.0252] [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] [Indexed: 10/05/2024] Open
Abstract
This review explores the regenerative potential of key progenitor cell types and therapeutic strategies to improve healing of complex fractures and bone defects. We define, summarize, and discuss the differentiation potential of totipotent, pluripotent, and multipotent stem cells, emphasizing the advantages and shortcomings of cell therapy for bone repair and regeneration. The fundamental role of mesenchymal stem cells is highlighted due to their multipotency to differentiate into the key lineage cells including osteoblasts, osteocytes, and chondrocytes, which are crucial for bone formation and remodeling. Hematopoietic stem cells (HSCs) also play a significant role; immune cells such as macrophages and T-cells modulate inflammation and tissue repair. Osteoclasts are multinucleated cells that are important to bone remodeling. Vascular progenitor (VP) cells are critical to oxygen and nutrient supply. The dynamic interplay among these lineages and their microenvironment is essential for effective bone restoration. Therapies involving cells that are more than "minimally manipulated" are controversial and include embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). ESCs, derived from early-stage embryos, possess pluripotent capabilities and have shown promise in preclinical studies for bone healing. iPSCs, reprogrammed from somatic cells, offer personalized medicine applications and can differentiate into various tissue-specific cell lines. Minimally manipulative cell therapy approaches such as the use of bone marrow aspirate concentrate (BMAC), exosomes, and various biomaterials for local delivery are explored for their effectiveness in bone regeneration. BMAC, which contains mostly immune cells but few mesenchymal and VPs, probably improves bone healing by facilitating paracrine-mediated intercellular communication. Exosome isolation harnesses the biological signals and cellular by-products that are a primary source for cell crosstalk and activation. Safe, efficacious, and cost-effective strategies to enhance bone healing using novel cellular therapies are part of a changing paradigm to modulate the inflammatory, repair, and regenerative pathways to achieve earlier more robust tissue healing and improved physical function. Impact Statement Stem cell therapy holds immense potential for bone healing due to its ability to regenerate damaged tissue. Nonmanipulated bone marrow aspirate contains mesenchymal stem cells that promote bone repair and reduce healing time. Induced pluripotent stem cells offer the advantage of creating patient-specific cells that can differentiate into osteoblasts, aiding in bone regeneration. Other delivery methods, such as scaffold-based techniques, enhance stem cell integration and function. Collectively, these approaches can improve treatment outcomes, reduce recovery periods, and advance our understanding of bone healing mechanisms, making them pivotal in orthopedic research and regenerative medicine.
Collapse
Affiliation(s)
- Simon Kwoon-Ho Chow
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Alexa Pius
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Mayu Morita
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Yasemin Ergul
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Masatoshi Murayama
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Issei Shinohara
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Mehmet Sertac Cekuc
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Chao Ma
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Yosuke Susuki
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
- Department of Bioengineering, Stanford University, Stanford, California, USA
| |
Collapse
|
9
|
Silveira ASDA, Alves ACDA, Gimenes GM, Quessada PDS, Lobato TB, Dias BB, Pereira ACG, Iser-Bem PN, Pereira JNB, Hatanaka E, Masi LN, Pithon-Curi TC, Mattaraia VGDM, Hirabara SM, Crisma AR, Gorjão R, Curi R. Evidence for a Pro-Inflammatory State of Macrophages from Non-Obese Type-2 Diabetic Goto-Kakizaki Rats. Int J Mol Sci 2024; 25:10240. [PMID: 39408569 PMCID: PMC11477416 DOI: 10.3390/ijms251910240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/07/2024] [Accepted: 09/13/2024] [Indexed: 10/20/2024] Open
Abstract
Obesity causes insulin resistance (IR) through systemic low-grade inflammation and can lead to type 2 diabetes mellitus (T2DM). However, the mechanisms that cause IR and T2DM in non-obese individuals are unclear. The Goto-Kakizaki (GK) rat develops IR spontaneously and is a model of non-obese T2DM. These rats exhibit hyperglycemia beginning at weaning and exhibit lower body mass than control Wistar rats. Herein, we tested the hypothesis that macrophages of GK rats are permanently in a pro-inflammatory state, which may be associated with a systemic inflammation condition that mimics the pathogenesis of obesity-induced T2DM. Using eighteen-week-old GK and control Wistar rats, we investigated the proportions of M1 (pro-inflammatory) and M2 (anti-inflammatory) macrophages isolated from the peritoneal cavity. Additionally, the production of inflammatory cytokines and reactive oxygen species (ROS) in cultured macrophages under basal and stimulated conditions was assessed. It was found that phorbol myristate acetate (PMA) stimulation increased GK rat macrophage ROS production 90-fold compared to basal levels. This response was also three times more pronounced than in control cells (36-fold). The production of pro-inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α), tended to be upregulated in cultured macrophages from GK rats under basal conditions. Macrophages from GK rats produced 1.6 times more granulocyte-macrophage colony-stimulating factor (GM-CSF), 1.5 times more monocyte chemoattractant protein-1 (MCP-1) and 3.3 times more TNF-α than control cells when stimulated with lipopolysaccharide (LPS) (p = 0.0033; p = 0.049; p = 0.002, respectively). Moreover, compared to control cells, GK rats had 60% more M1 (p = 0.0008) and 23% less M2 (p = 0.038) macrophages. This study is the first to report macrophage inflammatory reprogramming towards a pro-inflammatory state in GK rats.
Collapse
Affiliation(s)
- Amanda Santos de Almeida Silveira
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo 01506-000, SP, Brazil (T.B.L.); (S.M.H.)
| | - Amara Cassandra dos Anjos Alves
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo 01506-000, SP, Brazil (T.B.L.); (S.M.H.)
| | - Gabriela Mandú Gimenes
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo 01506-000, SP, Brazil (T.B.L.); (S.M.H.)
| | - Patrícia da Silva Quessada
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo 01506-000, SP, Brazil (T.B.L.); (S.M.H.)
| | - Tiago Bertola Lobato
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo 01506-000, SP, Brazil (T.B.L.); (S.M.H.)
| | - Beatriz Belmiro Dias
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo 01506-000, SP, Brazil (T.B.L.); (S.M.H.)
| | - Ana Carolina Gomes Pereira
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo 01506-000, SP, Brazil (T.B.L.); (S.M.H.)
| | - Patrícia Nancy Iser-Bem
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo 01506-000, SP, Brazil (T.B.L.); (S.M.H.)
| | - Joice Naiara Bertaglia Pereira
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo 01506-000, SP, Brazil (T.B.L.); (S.M.H.)
- Butantan Institute, São Paulo 05585-000, SP, Brazil
| | - Elaine Hatanaka
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo 01506-000, SP, Brazil (T.B.L.); (S.M.H.)
| | - Laureane Nunes Masi
- Multicenter Graduate Program in Physiological Sciences, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, SC 88037-000, Brazil
| | - Tânia Cristina Pithon-Curi
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo 01506-000, SP, Brazil (T.B.L.); (S.M.H.)
| | | | - Sandro Massao Hirabara
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo 01506-000, SP, Brazil (T.B.L.); (S.M.H.)
| | - Amanda Rabello Crisma
- Department of Clinical Analysis, Federal University of Paraná, Curitiba 80210-170, PR, Brazil
| | - Renata Gorjão
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo 01506-000, SP, Brazil (T.B.L.); (S.M.H.)
| | - Rui Curi
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo 01506-000, SP, Brazil (T.B.L.); (S.M.H.)
- Butantan Institute, São Paulo 05585-000, SP, Brazil
| |
Collapse
|
10
|
Fernandes CJC, Silva RA, Ferreira MR, Fuhler GM, Peppelenbosch MP, van der Eerden BC, Zambuzzi WF. Vascular smooth muscle cell-derived exosomes promote osteoblast-to-osteocyte transition via β-catenin signaling. Exp Cell Res 2024; 442:114211. [PMID: 39147261 DOI: 10.1016/j.yexcr.2024.114211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/01/2024] [Accepted: 08/13/2024] [Indexed: 08/17/2024]
Abstract
Blood vessel growth and osteogenesis in the skeletal system are coupled; however, fundamental aspects of vascular function in osteoblast-to-osteocyte transition remain unclear. Our study demonstrates that vascular smooth muscle cells (VSMCs), but not endothelial cells, are sufficient to drive bone marrow mesenchymal stromal cell-derived osteoblast-to-osteocyte transition via β-catenin signaling and exosome-mediated communication. We found that VSMC-derived exosomes are loaded with transcripts encoding proteins associated with the osteocyte phenotype and members of the WNT/β-catenin signaling pathway. In contrast, endothelial cell-derived exosomes facilitated mature osteoblast differentiation by reprogramming the TGFB1 gene family and osteogenic transcription factors osterix (SP7) and RUNX2. Notably, VSMCs express significant levels of tetraspanins (CD9, CD63, and CD81) and drive the intracellular trafficking of exosomes with a lower membrane zeta potential than those from other cells. Additionally, the high ATP content within these exosomes supports mineralization mechanisms, as ATP is a substrate for alkaline phosphatase. Osteocyte function was further validated by RNA sequencing, revealing activity in genes related to intermittent mineralization and sonic hedgehog signaling, alongside a significant increase in TNFSF11 levels. Our findings unveil a novel role of VSMCs in promoting osteoblast-to-osteocyte transition, thus offering new insights into bone biology and homeostasis, as well as in bone-related diseases. Clinically, these insights could pave the way for innovative therapeutic strategies targeting VSMC-derived exosome pathways to treat bone-related disorders such as osteoporosis. By manipulating these signaling pathways, it may be possible to enhance bone regeneration and improve skeletal health in patients with compromised bone structure and function.
Collapse
Affiliation(s)
- Célio J C Fernandes
- Bioassays and Cell Dynamics Lab, Dept. of Chemistry and Biochemistry, Bioscience Institute, UNESP, Botucatu, 18603-100, Sao Paulo, Brazil
| | - Rodrigo A Silva
- School of Dentistry, University of Taubaté, 12020-340, Taubaté, São Paulo, Brazil
| | - Marcel R Ferreira
- Bioassays and Cell Dynamics Lab, Dept. of Chemistry and Biochemistry, Bioscience Institute, UNESP, Botucatu, 18603-100, Sao Paulo, Brazil
| | - Gwenny M Fuhler
- Department of Gastroenterology and Hepatology, Erasmus MC, Erasmus University Medical Center, Dr Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC, Erasmus University Medical Center, Dr Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Bram Cj van der Eerden
- Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Dr Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Willian F Zambuzzi
- Bioassays and Cell Dynamics Lab, Dept. of Chemistry and Biochemistry, Bioscience Institute, UNESP, Botucatu, 18603-100, Sao Paulo, Brazil.
| |
Collapse
|
11
|
Bal Z, Takakura N. Hydrogel Use in Osteonecrosis of the Femoral Head. Gels 2024; 10:544. [PMID: 39195073 DOI: 10.3390/gels10080544] [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: 06/10/2024] [Revised: 07/11/2024] [Accepted: 07/20/2024] [Indexed: 08/29/2024] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a vascular disease of unknown etiology and can be categorized mainly into two types: non-traumatic and traumatic ONFH. Thus, understanding osteogenic-angiogenic coupling is of prime importance in finding a solution for the treatment of ONFH. Hydrogels are biomaterials that are similar to the extracellular matrix (ECM). As they are able to mimic real tissue, they meet one of the most important rules in tissue engineering. In ONFH studies, hydrogels have recently become popular because of their ability to retain water and their adjustable properties, injectability, and mimicry of natural ECM. Because bone regeneration and graft materials are very broad areas of research and ONFH is a complex situation including bone and vascular systems, and there is no settled treatment strategy for ONFH worldwide, in this review paper, we followed a top-down approach by reviewing (1) bone and bone grafting, (2) hydrogels, (3) vascular systems, and (4) ONFH and hydrogel use in ONFH with studies in the literature which show promising results in limited clinical studies. The aim of this review paper is to provide the reader with general information on every aspect of ONFH and to focus on the hydrogel used in ONFH.
Collapse
Affiliation(s)
- Zeynep Bal
- Laboratory of Signal Transduction, WPI Immunology Frontier Research Center (WPI-IFReC), Osaka University, 3-1 Yamada-oka, Suita 565-0871, Osaka, Japan
- Department of Signal Transduction, Research Institute for Microbial Diseases (RIMD), Osaka University, 3-1 Yamada-oka, Suita 565-0871, Osaka, Japan
| | - Nobuyuki Takakura
- Laboratory of Signal Transduction, WPI Immunology Frontier Research Center (WPI-IFReC), Osaka University, 3-1 Yamada-oka, Suita 565-0871, Osaka, Japan
- Department of Signal Transduction, Research Institute for Microbial Diseases (RIMD), Osaka University, 3-1 Yamada-oka, Suita 565-0871, Osaka, Japan
| |
Collapse
|
12
|
Yuan X, Zhu W, Yang Z, He N, Chen F, Han X, Zhou K. Recent Advances in 3D Printing of Smart Scaffolds for Bone Tissue Engineering and Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403641. [PMID: 38861754 DOI: 10.1002/adma.202403641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/15/2024] [Indexed: 06/13/2024]
Abstract
The repair and functional reconstruction of bone defects resulting from severe trauma, surgical resection, degenerative disease, and congenital malformation pose significant clinical challenges. Bone tissue engineering (BTE) holds immense potential in treating these severe bone defects, without incurring prevalent complications associated with conventional autologous or allogeneic bone grafts. 3D printing technology enables control over architectural structures at multiple length scales and has been extensively employed to process biomimetic scaffolds for BTE. In contrast to inert and functional bone grafts, next-generation smart scaffolds possess a remarkable ability to mimic the dynamic nature of native extracellular matrix (ECM), thereby facilitating bone repair and regeneration. Additionally, they can generate tailored and controllable therapeutic effects, such as antibacterial or antitumor properties, in response to exogenous and/or endogenous stimuli. This review provides a comprehensive assessment of the progress of 3D-printed smart scaffolds for BTE applications. It begins with an introduction to bone physiology, followed by an overview of 3D printing technologies utilized for smart scaffolds. Notable advances in various stimuli-responsive strategies, therapeutic efficacy, and applications of 3D-printed smart scaffolds are discussed. Finally, the review highlights the existing challenges in the development and clinical implementation of smart scaffolds, as well as emerging technologies in this field.
Collapse
Affiliation(s)
- Xun Yuan
- National Engineering Research Centre for High Efficiency Grinding, College of Mechanical and Vehicle Engineering, Hunan University, Changsha, 410082, China
| | - Wei Zhu
- National Engineering Research Centre for High Efficiency Grinding, College of Mechanical and Vehicle Engineering, Hunan University, Changsha, 410082, China
| | - Zhongyuan Yang
- National Engineering Research Centre for High Efficiency Grinding, College of Mechanical and Vehicle Engineering, Hunan University, Changsha, 410082, China
| | - Ning He
- National Engineering Research Centre for High Efficiency Grinding, College of Mechanical and Vehicle Engineering, Hunan University, Changsha, 410082, China
| | - Feng Chen
- National Engineering Research Centre for High Efficiency Grinding, College of Mechanical and Vehicle Engineering, Hunan University, Changsha, 410082, China
| | - Xiaoxiao Han
- National Engineering Research Centre for High Efficiency Grinding, College of Mechanical and Vehicle Engineering, Hunan University, Changsha, 410082, China
| | - Kun Zhou
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| |
Collapse
|
13
|
Trompet D, Melis S, Chagin AS, Maes C. Skeletal stem and progenitor cells in bone development and repair. J Bone Miner Res 2024; 39:633-654. [PMID: 38696703 DOI: 10.1093/jbmr/zjae069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/11/2024] [Accepted: 04/17/2024] [Indexed: 05/04/2024]
Abstract
Bone development, growth, and repair are complex processes involving various cell types and interactions, with central roles played by skeletal stem and progenitor cells. Recent research brought new insights into the skeletal precursor populations that mediate intramembranous and endochondral bone development. Later in life, many of the cellular and molecular mechanisms determining development are reactivated upon fracture, with powerful trauma-induced signaling cues triggering a variety of postnatal skeletal stem/progenitor cells (SSPCs) residing near the bone defect. Interestingly, in this injury context, the current evidence suggests that the fates of both SSPCs and differentiated skeletal cells can be considerably flexible and dynamic, and that multiple cell sources can be activated to operate as functional progenitors generating chondrocytes and/or osteoblasts. The combined implementation of in vivo lineage tracing, cell surface marker-based cell selection, single-cell molecular analyses, and high-resolution in situ imaging has strongly improved our insights into the diversity and roles of developmental and reparative stem/progenitor subsets, while also unveiling the complexity of their dynamics, hierarchies, and relationships. Albeit incompletely understood at present, findings supporting lineage flexibility and possibly plasticity among sources of osteogenic cells challenge the classical dogma of a single primitive, self-renewing, multipotent stem cell driving bone tissue formation and regeneration from the apex of a hierarchical and strictly unidirectional differentiation tree. We here review the state of the field and the newest discoveries in the origin, identity, and fates of skeletal progenitor cells during bone development and growth, discuss the contributions of adult SSPC populations to fracture repair, and reflect on the dynamism and relationships among skeletal precursors and differentiated cell lineages. Further research directed at unraveling the heterogeneity and capacities of SSPCs, as well as the regulatory cues determining their fate and functioning, will offer vital new options for clinical translation toward compromised fracture healing and bone regenerative medicine.
Collapse
Affiliation(s)
- Dana Trompet
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, 40530 Gothenburg, Sweden
- Department of Physiology and Pharmacology, Karolinska Institute, 17177 Stockholm, Sweden
| | - Seppe Melis
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Andrei S Chagin
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, 40530 Gothenburg, Sweden
- Department of Physiology and Pharmacology, Karolinska Institute, 17177 Stockholm, Sweden
| | - Christa Maes
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
14
|
Martini S, Drzeniek NM, Stark R, Kollert MR, Du W, Reinke S, Ort M, Hardt S, Kotko I, Kath J, Schlickeiser S, Geißler S, Wagner DL, Krebs AC, Volk HD. Long-term in vitromaintenance of plasma cells in a hydrogel-enclosed human bone marrow microphysiological 3D model system. Biofabrication 2024; 16:045005. [PMID: 38955197 DOI: 10.1088/1758-5090/ad5dfe] [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: 10/16/2023] [Accepted: 07/01/2024] [Indexed: 07/04/2024]
Abstract
Plasma cells (PCs) in bone marrow (BM) play an important role in both protective and pathogenic humoral immune responses, e.g. in various malignant and non-malignant diseases such as multiple myeloma, primary and secondary immunodeficiencies and autoimmune diseases. Dedicated microenvironmental niches in the BM provide PCs with biomechanical and soluble factors that support their long-term survival. There is a high need for appropriate and robust model systems to better understand PCs biology, to develop new therapeutic strategies for PCs-related diseases and perform targeted preclinical studies with high predictive value. Most preclinical data have been derived fromin vivostudies in mice, asin vitrostudies of human PCs are limited due to restricted survival and functionality in conventional 2D cultures that do not reflect the unique niche architecture of the BM. We have developed a microphysiological, dynamic 3D BM culture system (BM-MPS) based on human primary tissue (femoral biopsies), mechanically supported by a hydrogel scaffold casing. While a bioinert agarose casing did not support PCs survival, a photo-crosslinked collagen-hyaluronic acid (Col-HA) hydrogel preserved the native BM niche architecture and allowed PCs survivalin vitrofor up to 2 weeks. Further, the Col-HA hydrogel was permissive to lymphocyte migration into the microphysiological system´s circulation. Long-term PCs survival was related to the stable presence in the culture of soluble factors, as APRIL, BAFF, and IL-6. Increasing immunoglobulins concentrations in the medium confirm their functionality over culture time. To the best of our knowledge, this study is the first report of successful long-term maintenance of primary-derived non-malignant PCsin vitro. Our innovative model system is suitable for in-depthin vitrostudies of human PCs regulation and exploration of targeted therapeutic approaches such as CAR-T cell therapy or biologics.
Collapse
Affiliation(s)
- Stefania Martini
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Institute of Medical Immunology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Norman Michael Drzeniek
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Institute of Medical Immunology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Regina Stark
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Matthias Reiner Kollert
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Julius Wolff Institute, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Weijie Du
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Simon Reinke
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Melanie Ort
- Julius Wolff Institute, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Sebastian Hardt
- Center for Musculoskeletal Surgery Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Iuliia Kotko
- Institute of Medical Immunology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Jonas Kath
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Stephan Schlickeiser
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- CheckImmune GmbH, Berlin, Germany
| | - Sven Geißler
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Julius Wolff Institute, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Dimitrios Laurin Wagner
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- Institute of Transfusion Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Anna-Catharina Krebs
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Institute of Medical Immunology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Hans-Dieter Volk
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Institute of Medical Immunology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
- Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
15
|
Abadeer AI, Wu CM, Brooks DM, Higgins JP, Giladi AM, Shubinets V. Arterial Perfusion of the Proximal Phalanx Revisited: New Insights Based on Micro-Computed Tomography. J Hand Surg Am 2024; 49:713.e1-713.e7. [PMID: 36333245 DOI: 10.1016/j.jhsa.2022.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 09/08/2022] [Accepted: 09/21/2022] [Indexed: 11/06/2022]
Abstract
PURPOSE To characterize the periosteal and endosteal arterial perfusion of the proximal phalanx using micro-computed tomography angiography (micro-CTA). METHODS Cadaveric upper extremities were injected with a barium sulfate/gelatin suspension. Phalanges were imaged using micro-CTA and analyzed with a focus on osseous arterial anatomy. Periosteal and endosteal perfusion was characterized by number of vessels, length, anatomic course, and caliber. RESULTS The base of the proximal phalanx had a significantly greater number (8.0 ± 3.5) of periosteal vessels than those of the shaft (4.1 ± 1.6) and head (1.3 ± 1.1). One-third (34.4%) of the specimens demonstrated a complete absence of periosteal vessels in the head. A nutrient endosteal vessel was noted in 100% of the specimens. Entering at the junction of the middle and distal third of the bone (25.8 ± 3.9 mm from base), the nutrient vessel entered the proximal phalanx of the index finger along its ulnar aspect (8 of 8 specimens), the middle finger along its radial aspect (6 of 8), the ring finger along its ulnar aspect (5 of 8), and the little finger along its radial aspect (7 of 8). The nutrient vessel branched into proximal and distal extensions toward the shaft and head, respectively, with an average endosteal length of 10.7 ± 5.2 mm and average diameter of 0.36 ± 0.11 mm. CONCLUSIONS Periosteal contributions to the perfusion of the proximal phalanx appear to diminish distally. The endosteal arterial anatomy remains consistent, with a single nutrient vessel entering the intramedullary canal with reliable laterality on each digit. This is often the only vessel supplying the head of the proximal phalanx, making this area particularly susceptible to vascular compromise. CLINICAL RELEVANCE An understanding of the patterns of perfusion of the proximal phalanx provides some insight into clinically observed pathology, as well as guidance for operative management.
Collapse
Affiliation(s)
- Andrew I Abadeer
- The Curtis National Hand Center, Medstar Union Memorial Hospital, Baltimore, MD; Georgetown University School of Medicine, Washington, DC
| | - Caroline M Wu
- The Curtis National Hand Center, Medstar Union Memorial Hospital, Baltimore, MD; Georgetown University School of Medicine, Washington, DC
| | - Daina M Brooks
- Musculoskeletal Research Center, Department of Orthopedic Surgery, MedStar Union Memorial Hospital, Baltimore, MD
| | - James P Higgins
- The Curtis National Hand Center, Medstar Union Memorial Hospital, Baltimore, MD
| | - Aviram M Giladi
- The Curtis National Hand Center, Medstar Union Memorial Hospital, Baltimore, MD
| | - Valeriy Shubinets
- The Curtis National Hand Center, Medstar Union Memorial Hospital, Baltimore, MD.
| |
Collapse
|
16
|
Fleischhacker V, Milosic F, Bricelj M, Kührer K, Wahl‐Figlash K, Heimel P, Diendorfer A, Nardini E, Fischer I, Stangl H, Pietschmann P, Hackl M, Foisner R, Grillari J, Hengstschläger M, Osmanagic‐Myers S. Aged-vascular niche hinders osteogenesis of mesenchymal stem cells through paracrine repression of Wnt-axis. Aging Cell 2024; 23:e14139. [PMID: 38578073 PMCID: PMC11166365 DOI: 10.1111/acel.14139] [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: 07/21/2023] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 04/06/2024] Open
Abstract
Age-induced decline in osteogenic potential of bone marrow mesenchymal stem cells (BMSCs) potentiates osteoporosis and increases the risk for bone fractures. Despite epidemiology studies reporting concurrent development of vascular and bone diseases in the elderly, the underlying mechanisms for the vascular-bone cross-talk in aging are largely unknown. In this study, we show that accelerated endothelial aging deteriorates bone tissue through paracrine repression of Wnt-driven-axis in BMSCs. Here, we utilize physiologically aged mice in conjunction with our transgenic endothelial progeria mouse model (Hutchinson-Gilford progeria syndrome; HGPS) that displays hallmarks of an aged bone marrow vascular niche. We find bone defects associated with diminished BMSC osteogenic differentiation that implicate the existence of angiocrine factors with long-term inhibitory effects. microRNA-transcriptomics of HGPS patient plasma combined with aged-vascular niche analyses in progeria mice reveal abundant secretion of Wnt-repressive microRNA-31-5p. Moreover, we show that inhibition of microRNA-31-5p as well as selective Wnt-activator CHIR99021 boosts the osteogenic potential of BMSCs through de-repression and activation of the Wnt-signaling, respectively. Our results demonstrate that the vascular niche significantly contributes to osteogenesis defects in aging and pave the ground for microRNA-based therapies of bone loss in elderly.
Collapse
Affiliation(s)
| | - Filip Milosic
- Center for Pathobiochemistry and GeneticsMedical University of ViennaViennaAustria
| | - Marko Bricelj
- Center for Pathobiochemistry and GeneticsMedical University of ViennaViennaAustria
| | - Kristina Kührer
- Center for Pathobiochemistry and GeneticsMedical University of ViennaViennaAustria
| | - Katharina Wahl‐Figlash
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
| | - Patrick Heimel
- Ludwig Boltzmann Institute for Traumatology (The Research Center in Cooperation with AUVA)ViennaAustria
- Austrian Cluster for Tissue RegenerationViennaAustria
- Core Facility Hard Tissue and Biomaterial Research, Karl Donath LaboratoryUniversity Clinic of Dentistry, Medical University of ViennaViennaAustria
| | | | - Eleonora Nardini
- Center for Pathobiochemistry and GeneticsMedical University of ViennaViennaAustria
| | | | - Herbert Stangl
- Center for Pathobiochemistry and GeneticsMedical University of ViennaViennaAustria
| | - Peter Pietschmann
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
| | | | - Roland Foisner
- Max Perutz Labs, Vienna BioCenter Campus (VBC)ViennaAustria
- Max Perutz LabsMedical University of ViennaViennaAustria
| | - Johannes Grillari
- Ludwig Boltzmann Institute for Traumatology (The Research Center in Cooperation with AUVA)ViennaAustria
- Austrian Cluster for Tissue RegenerationViennaAustria
- Department of Biotechnology, Institute of Molecular BiotechnologyUniversity of Natural Resources and Life SciencesViennaAustria
| | | | | |
Collapse
|
17
|
Wang Q, Peng F, Yang J, Chen X, Peng Z, Zhang M, Tang D, Liu J, Zhao H. MicroRNAs regulate the vicious cycle of vascular calcification-osteoporosis in postmenopausal women. Mol Biol Rep 2024; 51:622. [PMID: 38709309 DOI: 10.1007/s11033-024-09550-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/12/2024] [Indexed: 05/07/2024]
Abstract
Menopause is a normal physiological process accompanied by changes in various physiological states. The incidence of vascular calcification (VC) increases each year after menopause and is closely related to osteoporosis (OP). Although many studies have investigated the links between VC and OP, the interaction mechanism of the two under conditions of estrogen loss remains unclear. MicroRNAs (miRNAs), which are involved in epigenetic modification, play a critical role in estrogen-mediated mineralization. In the past several decades, miRNAs have been identified as biomarkers or therapeutic targets in diseases. Thus, we hypothesize that these small molecules can provide new diagnostic and therapeutic approaches. In this review, we summarize the close interactions between VC and OP and the role of miRNAs in their interplay.
Collapse
Affiliation(s)
- Qian Wang
- Department of Radiology, The First Affiliated Hospital of The University of South China, Hengyang, Hunan, China
| | - Fei Peng
- Department of Radiology, The First Affiliated Hospital of The University of South China, Hengyang, Hunan, China
| | - Jing Yang
- Changsha Central Hospital Affiliated to University of South China, Changsha, Hunan, China
| | - Xiaolong Chen
- Department of Radiology, The First Affiliated Hospital of The University of South China, Hengyang, Hunan, China
| | - Zhaojie Peng
- Department of Radiology, The First Affiliated Hospital of The University of South China, Hengyang, Hunan, China
| | - Minyi Zhang
- The University of South China, Hengyang, Hunan, China
| | - Deqiu Tang
- Department of Radiology, The First Affiliated Hospital of The University of South China, Hengyang, Hunan, China
| | - Jianghua Liu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of The University of South China, Hengyang, Hunan, China.
| | - Heng Zhao
- Department of Radiology, The First Affiliated Hospital of The University of South China, Hengyang, Hunan, China.
| |
Collapse
|
18
|
Tang J, Wang X, Lin X, Wu C. Mesenchymal stem cell-derived extracellular vesicles: a regulator and carrier for targeting bone-related diseases. Cell Death Discov 2024; 10:212. [PMID: 38697996 PMCID: PMC11066013 DOI: 10.1038/s41420-024-01973-w] [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: 02/05/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 05/05/2024] Open
Abstract
The escalating threat of bone-related diseases poses a significant challenge to human health. Mesenchymal stem cell (MSC)-derived extracellular vesicles (MSC-EVs), as inherent cell-secreted natural products, have emerged as promising treatments for bone-related diseases. Leveraging outstanding features such as high biocompatibility, low immunogenicity, superior biological barrier penetration, and extended circulating half-life, MSC-EVs serve as potent carriers for microRNAs (miRNAs), long no-code RNAs (lncRNAs), and other biomolecules. These cargo molecules play pivotal roles in orchestrating bone metabolism and vascularity through diverse mechanisms, thereby contributing to the amelioration of bone diseases. Additionally, engineering modifications enhance the bone-targeting ability of MSC-EVs, mitigating systemic side effects and bolstering their clinical translational potential. This review comprehensively explores the mechanisms through which MSC-EVs regulate bone-related disease progression. It delves into the therapeutic potential of MSC-EVs as adept drug carriers, augmented by engineered modification strategies tailored for osteoarthritis (OA), rheumatoid arthritis (RA), osteoporosis, and osteosarcoma. In conclusion, the exceptional promise exhibited by MSC-EVs positions them as an excellent solution with considerable translational applications in clinical orthopedics.
Collapse
Affiliation(s)
- Jiandong Tang
- Orthopaedics Center, Zigong Fourth People's Hospital, Tan mu lin Street 19#, Zigong, 643099, Sichuan Province, China
| | - Xiangyu Wang
- Orthopaedics Center, Zigong Fourth People's Hospital, Tan mu lin Street 19#, Zigong, 643099, Sichuan Province, China
| | - Xu Lin
- Orthopaedics Center, Zigong Fourth People's Hospital, Tan mu lin Street 19#, Zigong, 643099, Sichuan Province, China
| | - Chao Wu
- Orthopaedics Center, Zigong Fourth People's Hospital, Tan mu lin Street 19#, Zigong, 643099, Sichuan Province, China.
| |
Collapse
|
19
|
Fan Y, Zhang W, Huang X, Fan M, Shi C, Zhao L, Pi G, Zhang H, Ni S. Senescent-like macrophages mediate angiogenesis for endplate sclerosis via IL-10 secretion in male mice. Nat Commun 2024; 15:2939. [PMID: 38580630 PMCID: PMC10997778 DOI: 10.1038/s41467-024-47317-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 03/25/2024] [Indexed: 04/07/2024] Open
Abstract
Endplate sclerosis is a notable aspect of spine degeneration or aging, but the mechanisms remain unclear. Here, we report that senescent macrophages accumulate in the sclerotic endplates of lumbar spine instability (LSI) or aging male mouse model. Specifically, knockout of cdkn2a (p16) in macrophages abrogates LSI or aging-induced angiogenesis and sclerosis in the endplates. Furthermore, both in vivo and in vitro studies indicate that IL-10 is the primary elevated cytokine of senescence-related secretory phenotype (SASP). Mechanistically, IL-10 increases pSTAT3 in endothelial cells, leading to pSTAT3 directly binding to the promoters of Vegfa, Mmp2, and Pdgfb to encourage their production, resulting in angiogenesis. This study provides information on understanding the link between immune senescence and endplate sclerosis, which might be useful for therapeutic approaches.
Collapse
Affiliation(s)
- Yonggang Fan
- Department of Orthopaedics, 1st Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, PR China
| | - Weixin Zhang
- Zhejiang Chinese Medicine University, Hangzhou, 310053, PR China
| | - Xiusheng Huang
- Department of Orthopaedics, 1st Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, PR China
| | - Mingzhe Fan
- Department of Orthopaedics, 1st Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, PR China
| | - Chenhao Shi
- Department of Orthopaedics, 1st Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, PR China
| | - Lantian Zhao
- Department of Orthopaedics, 1st Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, PR China
| | - Guofu Pi
- Department of Orthopaedics, 1st Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, PR China
| | - Huafeng Zhang
- Department of Orthopaedics, 1st Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, PR China
| | - Shuangfei Ni
- Department of Orthopaedics, 1st Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, PR China.
| |
Collapse
|
20
|
Dong Q, Fei X, Zhang H, Zhu X, Ruan J. Effect of Dimethyloxalylglycine on Stem Cells Osteogenic Differentiation and Bone Tissue Regeneration-A Systematic Review. Int J Mol Sci 2024; 25:3879. [PMID: 38612687 PMCID: PMC11011423 DOI: 10.3390/ijms25073879] [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: 02/20/2024] [Revised: 03/20/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Dimethyloxalylglycine (DMOG) has been found to stimulate osteogenesis and angiogenesis of stem cells, promoting neo-angiogenesis in bone tissue regeneration. In this review, we conducted a comprehensive search of the literature to investigate the effects of DMOG on osteogenesis and bone regeneration. We screened the studies based on specific inclusion criteria and extracted relevant information from both in vitro and in vivo experiments. The risk of bias in animal studies was evaluated using the SYRCLE tool. Out of the 174 studies retrieved, 34 studies met the inclusion criteria (34 studies were analyzed in vitro and 20 studies were analyzed in vivo). The findings of the included studies revealed that DMOG stimulated stem cells' differentiation toward osteogenic, angiogenic, and chondrogenic lineages, leading to vascularized bone and cartilage regeneration. Addtionally, DMOG demonstrated therapeutic effects on bone loss caused by bone-related diseases. However, the culture environment in vitro is notably distinct from that in vivo, and the animal models used in vivo experiments differ significantly from humans. In summary, DMOG has the ability to enhance the osteogenic and angiogenic differentiation potential of stem cells, thereby improving bone regeneration in cases of bone defects. This highlights DMOG as a potential focus for research in the field of bone tissue regeneration engineering.
Collapse
Affiliation(s)
- Qiannan Dong
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710000, China
- Center of Oral Public Health, College of Stomatology, Xi’an Jiaotong University, Xi’an 710000, China
| | - Xiuzhi Fei
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710000, China
- Center of Oral Public Health, College of Stomatology, Xi’an Jiaotong University, Xi’an 710000, China
| | - Hengwei Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710000, China
- Center of Oral Public Health, College of Stomatology, Xi’an Jiaotong University, Xi’an 710000, China
| | - Ximei Zhu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710000, China
- Center of Oral Public Health, College of Stomatology, Xi’an Jiaotong University, Xi’an 710000, China
| | - Jianping Ruan
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710000, China
- Center of Oral Public Health, College of Stomatology, Xi’an Jiaotong University, Xi’an 710000, China
| |
Collapse
|
21
|
Manescu (Paltanea) V, Paltanea G, Antoniac A, Gruionu LG, Robu A, Vasilescu M, Laptoiu SA, Bita AI, Popa GM, Cocosila AL, Silviu V, Porumb A. Mechanical and Computational Fluid Dynamic Models for Magnesium-Based Implants. MATERIALS (BASEL, SWITZERLAND) 2024; 17:830. [PMID: 38399081 PMCID: PMC10890492 DOI: 10.3390/ma17040830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 01/26/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024]
Abstract
Today, mechanical properties and fluid flow dynamic analysis are considered to be two of the most important steps in implant design for bone tissue engineering. The mechanical behavior is characterized by Young's modulus, which must have a value close to that of the human bone, while from the fluid dynamics point of view, the implant permeability and wall shear stress are two parameters directly linked to cell growth, adhesion, and proliferation. In this study, we proposed two simple geometries with a three-dimensional pore network dedicated to a manufacturing route based on a titanium wire waving procedure used as an intermediary step for Mg-based implant fabrication. Implant deformation under different static loads, von Mises stresses, and safety factors were investigated using finite element analysis. The implant permeability was computed based on Darcy's law following computational fluid dynamic simulations and, based on the pressure drop, was numerically estimated. It was concluded that both models exhibited a permeability close to the human trabecular bone and reduced wall shear stresses within the biological range. As a general finding, the proposed geometries could be useful in orthopedics for bone defect treatment based on numerical analyses because they mimic the trabecular bone properties.
Collapse
Affiliation(s)
- Veronica Manescu (Paltanea)
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (A.R.); (M.V.); (S.A.L.)
- Faculty of Electrical Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania;
| | - Gheorghe Paltanea
- Faculty of Electrical Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania;
| | - Aurora Antoniac
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (A.R.); (M.V.); (S.A.L.)
| | - Lucian Gheorghe Gruionu
- Faculty of Mechanics, University of Craiova, 13 Alexandru Ioan Cuza, RO-200585 Craiova, Romania;
| | - Alina Robu
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (A.R.); (M.V.); (S.A.L.)
| | - Marius Vasilescu
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (A.R.); (M.V.); (S.A.L.)
| | - Stefan Alexandru Laptoiu
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (A.R.); (M.V.); (S.A.L.)
| | - Ana Iulia Bita
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (A.R.); (M.V.); (S.A.L.)
| | - Georgiana Maria Popa
- Department of Surgical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 10 P-ta 1 December Street, RO-410073 Oradea, Romania; (G.M.P.); (A.L.C.); (V.S.)
| | - Andreea Liliana Cocosila
- Department of Surgical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 10 P-ta 1 December Street, RO-410073 Oradea, Romania; (G.M.P.); (A.L.C.); (V.S.)
| | - Vlad Silviu
- Department of Surgical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 10 P-ta 1 December Street, RO-410073 Oradea, Romania; (G.M.P.); (A.L.C.); (V.S.)
| | - Anca Porumb
- Department of Dental Medicine, Faculty of Medicine and Pharmacy, University of Oradea, 10 P-ta 1 December Street, RO-410073 Oradea, Romania;
| |
Collapse
|
22
|
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
|
23
|
Hasan T, Pasala AR, Hassan D, Hanotaux J, Allan DS, Maganti HB. Homing and Engraftment of Hematopoietic Stem Cells Following Transplantation: A Pre-Clinical Perspective. Curr Oncol 2024; 31:603-616. [PMID: 38392038 PMCID: PMC10888387 DOI: 10.3390/curroncol31020044] [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: 12/22/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 02/24/2024] Open
Abstract
Hematopoietic stem-cell (HSC) transplantation (HSCT) is used to treat various hematologic disorders. Use of genetically modified mouse models of hematopoietic cell transplantation has been critical in our fundamental understanding of HSC biology and in developing approaches for human patients. Pre-clinical studies in animal models provide insight into the journey of transplanted HSCs from infusion to engraftment in bone-marrow (BM) niches. Various signaling molecules and growth factors secreted by HSCs and the niche microenvironment play critical roles in homing and engraftment of the transplanted cells. The sustained equilibrium of these chemical and biologic factors ensures that engrafted HSCs generate healthy and durable hematopoiesis. Transplanted healthy HSCs compete with residual host cells to repopulate stem-cell niches in the marrow. Stem-cell niches, in particular, can be altered by the effects of previous treatments, aging, and the paracrine effects of leukemic cells, which create inhospitable bone-marrow niches that are unfavorable for healthy hematopoiesis. More work to understand how stem-cell niches can be restored to favor normal hematopoiesis may be key to reducing leukemic relapses following transplant.
Collapse
Affiliation(s)
- Tanvir Hasan
- Canadian Blood Services, Stem Cells and Centre for Innovation, Ottawa, ON K1G 4J5, Canada; (T.H.); (A.R.P.); (D.H.); (J.H.)
| | - Ajay Ratan Pasala
- Canadian Blood Services, Stem Cells and Centre for Innovation, Ottawa, ON K1G 4J5, Canada; (T.H.); (A.R.P.); (D.H.); (J.H.)
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8L6, Canada
| | - Dhuha Hassan
- Canadian Blood Services, Stem Cells and Centre for Innovation, Ottawa, ON K1G 4J5, Canada; (T.H.); (A.R.P.); (D.H.); (J.H.)
| | - Justine Hanotaux
- Canadian Blood Services, Stem Cells and Centre for Innovation, Ottawa, ON K1G 4J5, Canada; (T.H.); (A.R.P.); (D.H.); (J.H.)
| | - David S. Allan
- Canadian Blood Services, Stem Cells and Centre for Innovation, Ottawa, ON K1G 4J5, Canada; (T.H.); (A.R.P.); (D.H.); (J.H.)
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8L6, Canada
- Clinical Epidemiology & Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, ON K1Y 4E9, Canada
| | - Harinad B. Maganti
- Canadian Blood Services, Stem Cells and Centre for Innovation, Ottawa, ON K1G 4J5, Canada; (T.H.); (A.R.P.); (D.H.); (J.H.)
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8L6, Canada
| |
Collapse
|
24
|
Picoli CDC, Birbrair A, Li Z. Pericytes as the Orchestrators of Vasculature and Adipogenesis. Genes (Basel) 2024; 15:126. [PMID: 38275607 PMCID: PMC10815550 DOI: 10.3390/genes15010126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
Pericytes (PCs) are located surrounding the walls of small blood vessels, particularly capillaries and microvessels. In addition to their functions in maintaining vascular integrity, participating in angiogenesis, and regulating blood flow, PCs also serve as a reservoir for multi-potent stem/progenitor cells in white, brown, beige, and bone marrow adipose tissues. Due to the complex nature of this cell population, the identification and characterization of PCs has been challenging. A comprehensive understanding of the heterogeneity of PCs may enhance their potential as therapeutic targets for metabolic syndromes or bone-related diseases. This mini-review summarizes multiple PC markers commonly employed in lineage-tracing studies, with an emphasis on their contribution to adipogenesis and functions in different adipose depots under diverse metabolic conditions.
Collapse
Affiliation(s)
| | - Alexander Birbrair
- Department of Dermatology, University of Wisconsin-Madison, Medical Sciences Center, Madison, WI 53706, USA;
| | - Ziru Li
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME 04074, USA;
| |
Collapse
|
25
|
Ye Z, Liu Y, Song J, Gao Y, Fang H, Hu Z, Zhang M, Liao W, Cui L, Liu Y. Expanding the therapeutic potential of Salvia miltiorrhiza: a review of its pharmacological applications in musculoskeletal diseases. Front Pharmacol 2023; 14:1276038. [PMID: 38116081 PMCID: PMC10728493 DOI: 10.3389/fphar.2023.1276038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023] Open
Abstract
Salvia miltiorrhiz, commonly known as "Danshen" in Chinese medicine, has longstanding history of application in cardiovascular and cerebrovascular diseases. Renowned for its diverse therapeutic properties, including promoting blood circulation, removing blood stasis, calming the mind, tonifying the blood, and benefiting the "Qi", recent studies have revealed its significant positive effects on bone metabolism. This potential has garnered attention for its promising role in treating musculoskeletal disorders. Consequently, there is a high anticipation for a comprehensive review of the potential of Salvia miltiorrhiza in the treatment of various musculoskeletal diseases, effectively introducing an established traditional Chinese medicine into a burgeoning field. AIM OF THE REVIEW Musculoskeletal diseases (MSDs) present significant challenges to healthcare systems worldwide. Previous studies have demonstrated the high efficacy and prospects of Salvia miltiorrhiza and its active ingredients for treatment of MSDs. This review aims to illuminate the newfound applications of Salvia miltiorrhiza and its active ingredients in the treatment of various MSDs, effectively bridging the gap between an established medicine and an emerging field. METHODS In this review, previous studies related to Salvia miltiorrhiza and its active ingredients on the treatment of MSD were collected, the specific active ingredients of Salvia miltiorrhiza were summarized, the effects of Salvia miltiorrhiza and its active ingredients for the treatment of MSDs, as well as their potential molecular mechanisms were reviewed and discussed. RESULTS Based on previous publications, Salvianolic acid A, salvianolic acid B, tanshinone IIA are the representative active ingredients of Salvia miltiorrhiza. Their application has shown significant beneficial outcomes in osteoporosis, fractures, and arthritis. Salvia miltiorrhiza and its active ingredients protect against MSDs by regulating different signaling pathways, including ROS, Wnt, MAPK, and NF-κB signaling. CONCLUSION Salvia miltiorrhiza and its active ingredients demonstrate promising potential for bone diseases and have been explored across a wide variety of MSDs. Further exploration of Salvia miltiorrhiza's pharmacological applications in MSDs holds great promise for advancing therapeutic interventions and improving the lives of patients suffering from these diseases.
Collapse
Affiliation(s)
- Zhiqiang Ye
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Provincial Administration of Traditional Chinese Medicine (Central People’s Hospital of Zhanjiang), Zhanjiang, China
| | - Yuyu Liu
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Jintong Song
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Provincial Administration of Traditional Chinese Medicine (Central People’s Hospital of Zhanjiang), Zhanjiang, China
| | - Yin Gao
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
- Marine Medical Research Institute of Zhanjiang, Zhanjiang, China
| | - Haiping Fang
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Zilong Hu
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Min Zhang
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Wenwei Liao
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Liao Cui
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
| | - Yanzhi Liu
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Provincial Administration of Traditional Chinese Medicine (Central People’s Hospital of Zhanjiang), Zhanjiang, China
| |
Collapse
|
26
|
Wu X, Zhai F, Chang A, Wei J, Guo Y, Zhang J. Application of machine learning algorithms to predict osteoporosis in postmenopausal women with type 2 diabetes mellitus. J Endocrinol Invest 2023; 46:2535-2546. [PMID: 37171784 DOI: 10.1007/s40618-023-02109-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 05/03/2023] [Indexed: 05/13/2023]
Abstract
PURPOSE The screening and diagnosis of osteoporosis in patients with type 2 diabetes mellitus (T2DM) based on bone mineral density remains challenging because of the limited availability and accessibility of dual-energy X-ray absorptiometry. We aimed to develop and validate models to predict the risk of osteoporosis in postmenopausal women with T2DM based on machine learning (ML) algorithms. METHODS This retrospective study included 303 postmenopausal women with T2DM. To develop prediction models for osteoporosis, we applied nine ML algorithms combined with demographic, clinical, and laboratory data. The least absolute shrinkage and selection operator were used to perform feature selection. We used the bootstrap resampling technique for model training and validation. To test the performance of the models, we calculated indices including the area under the receiver operating characteristic curve (AUROC), accuracy, sensitivity, specificity, positive predictive value, negative predictive value, F1 score, calibration curve, and decision curve analysis. Furthermore, we conducted fivefold cross-validation for parameter optimization and model validation. Feature importance was assessed using the SHapley additive explanation (SHAP). RESULTS We identified 10 independent predictors as the most valuable features. An AUROC of 0.616-1.000 was observed for nine ML algorithms. The extreme gradient boosting (XGBoost) model exhibited the best performance, outperforming conventional risk assessment tools and registering 0.993 in the training set, 0.798 in the validation set, and 0.786 in the test set for fivefold cross-validation. Using SHAP, we found that the explanatory variables contributed to the model and their relationship with osteoporosis occurrence. Furthermore, we developed a user-friendly tool for calculating the risk of osteoporosis. CONCLUSIONS With the integration of demographic and clinical risk factors, ML algorithms can accurately predict osteoporosis. The XGBoost model showed ideal performance. With the incorporation of these models in the clinic, patients may benefit from early osteoporosis diagnosis and treatment.
Collapse
Affiliation(s)
- X Wu
- Department of Endocrinology, Cangzhou Central Hospital, 16 Xinhua West Road, Cangzhou, 061000, Hebei, People's Republic of China.
| | - F Zhai
- Gynecological Clinic, Cangzhou Central Hospital, 16 Xinhua West Road, Cangzhou, 061000, Hebei, People's Republic of China
| | - A Chang
- Department of Endocrinology, Cangzhou Central Hospital, 16 Xinhua West Road, Cangzhou, 061000, Hebei, People's Republic of China
| | - J Wei
- Department of Endocrinology, Cangzhou Central Hospital, 16 Xinhua West Road, Cangzhou, 061000, Hebei, People's Republic of China
| | - Y Guo
- Department of Endocrinology, Cangzhou Central Hospital, 16 Xinhua West Road, Cangzhou, 061000, Hebei, People's Republic of China
| | - J Zhang
- Department of Endocrinology, Cangzhou Central Hospital, 16 Xinhua West Road, Cangzhou, 061000, Hebei, People's Republic of China
| |
Collapse
|
27
|
Zhang L, Guan Q, Wang Z, Feng J, Zou J, Gao B. Consequences of Aging on Bone. Aging Dis 2023; 15:2417-2452. [PMID: 38029404 PMCID: PMC11567267 DOI: 10.14336/ad.2023.1115] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/16/2023] [Indexed: 12/01/2023] Open
Abstract
With the aging of the global population, the incidence of musculoskeletal diseases has been increasing, seriously affecting people's health. As people age, the microenvironment within skeleton favors bone resorption and inhibits bone formation, accompanied by bone marrow fat accumulation and multiple cellular senescence. Specifically, skeletal stem/stromal cells (SSCs) during aging tend to undergo adipogenesis rather than osteogenesis. Meanwhile, osteoblasts, as well as osteocytes, showed increased apoptosis, decreased quantity, and multiple functional limitations including impaired mechanical sensing, intercellular modulation, and exosome secretion. Also, the bone resorption function of macrophage-lineage cells (including osteoclasts and preosteoclasts) was significantly enhanced, as well as impaired vascularization and innervation. In this study, we systematically reviewed the effect of aging on bone and the within microenvironment (including skeletal cells as well as their intracellular structure variations, vascular structures, innervation, marrow fat distribution, and lymphatic system) caused by aging, and mechanisms of osteoimmune regulation of the bone environment in the aging state, and the causal relationship with multiple musculoskeletal diseases in addition with their potential therapeutic strategy.
Collapse
Affiliation(s)
- Lingli Zhang
- College of Athletic Performance, Shanghai University of Sport, Shanghai, China
| | - Qiao Guan
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Zhikun Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Jie Feng
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Jun Zou
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Bo Gao
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
| |
Collapse
|
28
|
Liu X, Zhang P, Gu Y, Guo Q, Liu Y. Type H vessels: functions in bone development and diseases. Front Cell Dev Biol 2023; 11:1236545. [PMID: 38033859 PMCID: PMC10687371 DOI: 10.3389/fcell.2023.1236545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Type H vessels are specialized blood vessels found in the bone marrow that are closely associated with osteogenic activity. They are characterized by high expression of endomucin and CD31. Type H vessels form in the cancellous bone area during long bone development to provide adequate nutritional support for cells near the growth plate. They also influence the proliferation and differentiation of osteoprogenitors and osteoclasts in a paracrine manner, thereby creating a suitable microenvironment to facilitate new bone formation. Because of the close relationship between type H vessels and osteogenic activity, it has been found that type H vessels play a role in the physiological and pathological processes of bone diseases such as fracture healing, osteoporosis, osteoarthritis, osteonecrosis, and tumor bone metastasis. Moreover, experimental treatments targeting type H vessels can improve the outcomes of these diseases. Here, we reviewed the molecular mechanisms related to type H vessels and their associated osteogenic activities, which are helpful in further understanding the role of type H vessels in bone metabolism and will provide a theoretical basis and ideas for comprehending bone diseases from the vascular perspective.
Collapse
Affiliation(s)
- Xiaonan Liu
- Department of Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Colorectal and Anal Surgery, Zhongshan City People’s Hospital, Zhongshan, Guangdong, China
| | - Peilin Zhang
- Department of Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Gu
- Department of Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiaoyue Guo
- Endocrinology Research Center, Department of Endocrinology, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yonggan Liu
- Department of Colorectal and Anal Surgery, Zhongshan City People’s Hospital, Zhongshan, Guangdong, China
| |
Collapse
|
29
|
Campanile M, Bettinelli L, Cerutti C, Spinetti G. Bone marrow vasculature advanced in vitro models for cancer and cardiovascular research. Front Cardiovasc Med 2023; 10:1261849. [PMID: 37915743 PMCID: PMC10616801 DOI: 10.3389/fcvm.2023.1261849] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 09/12/2023] [Indexed: 11/03/2023] Open
Abstract
Cardiometabolic diseases and cancer are among the most common diseases worldwide and are a serious concern to the healthcare system. These conditions, apparently distant, share common molecular and cellular determinants, that can represent targets for preventive and therapeutic approaches. The bone marrow plays an important role in this context as it is the main source of cells involved in cardiovascular regeneration, and one of the main sites of liquid and solid tumor metastasis, both characterized by the cellular trafficking across the bone marrow vasculature. The bone marrow vasculature has been widely studied in animal models, however, it is clear the need for human-specific in vitro models, that resemble the bone vasculature lined by endothelial cells to study the molecular mechanisms governing cell trafficking. In this review, we summarized the current knowledge on in vitro models of bone marrow vasculature developed for cardiovascular and cancer research.
Collapse
Affiliation(s)
- Marzia Campanile
- Laboratory of Cardiovascular Research, IRCCS MultiMedica, Milan, Italy
| | - Leonardo Bettinelli
- Laboratory of Cardiovascular Research, IRCCS MultiMedica, Milan, Italy
- Department of Experimental Oncology, IRCCS-IEO, European Institute of Oncology, Milan, Italy
| | - Camilla Cerutti
- Department of Experimental Oncology, IRCCS-IEO, European Institute of Oncology, Milan, Italy
| | - Gaia Spinetti
- Laboratory of Cardiovascular Research, IRCCS MultiMedica, Milan, Italy
| |
Collapse
|
30
|
Jin L, Long Y, Zhang Q, Long J. MiRNAs regulate cell communication in osteogenesis-angiogenesis coupling during bone regeneration. Mol Biol Rep 2023; 50:8715-8728. [PMID: 37642761 DOI: 10.1007/s11033-023-08709-6] [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: 03/24/2023] [Accepted: 07/25/2023] [Indexed: 08/31/2023]
Abstract
Bone regeneration is a complex process that requires not only the participation of multiple cell types, but also signal communication between cells. The two basic processes of osteogenesis and angiogenesis are closely related to bone regeneration and bone homeostasis. H-type vessels are a subtype of bone vessels characterized by high expression of CD31 and EMCN. These vessels play a key role in the regulation of bone regeneration and are important mediators of coupling between osteogenesis and angiogenesis. Molecular regulation between different cell types is important for coordination of osteogenesis and angiogenesis that promotes bone regeneration. MiRNAs are small non-coding RNAs that predominantly regulate gene expression at the post-transcriptional level and are closely related to cell communication. Specifically, miRNAs transduce external stimuli through various cell signaling pathways and cause a series of physiological and pathological effects. They are also deeply involved in the bone repair process. This review focuses on three signaling pathways related to osteogenesis-angiogenesis coupling, as well as the miRNAs involved in these pathways. Elucidation of the molecular mechanisms governing osteogenesis and angiogenesis is of great significance for bone regeneration.
Collapse
Affiliation(s)
- Liangyu Jin
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, 610041, PR China
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu, 610041, PR China
- National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, PR China
| | - Yifei Long
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, 610041, PR China
- National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, PR China
| | - Qiuling Zhang
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, 610041, PR China
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu, 610041, PR China
- National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, PR China
| | - Jie Long
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, 610041, PR China.
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu, 610041, PR China.
- National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, PR China.
| |
Collapse
|
31
|
Wüster J, Hesse B, Rothweiler R, Bortel E, Gross C, Bakhtiyari S, King A, Boller E, Gerber J, Rendenbach C, Fretwurst T, Preissner S, Heiland M, Nelson K, Nahles S. Comparison of the 3D-microstructure of human alveolar and fibula bone in microvascular autologous bone transplantation: a synchrotron radiation μ-CT study. Front Bioeng Biotechnol 2023; 11:1169385. [PMID: 37691907 PMCID: PMC10486015 DOI: 10.3389/fbioe.2023.1169385] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 08/01/2023] [Indexed: 09/12/2023] Open
Abstract
Introduction: Autologous bone transplantation is successfully used in reconstructive surgery of large/critical-sized bone defects, whereby the microvascular free fibula flap is still regarded as the gold standard for the reconstruction of such defects in the head and neck region. Here, we report the morphological and lacunar properties of patient-paired bone samples from eight patients from the jaw (AB; recipient site) and the fibula (FB; donor site) on the micron length-scale using Synchrotron µ-CT. Insights into differences and similarities between these bone structures could offer a better understanding of the underlying mechanism for successful surgical outcomes and might clear the path for optimized, nature-inspired bone scaffold designs. Methods: Spatial vessel-pore arrangements, bone morphology, fluid-simulation derived permeability tensor, osteocyte lacunar density, and lacunar morphology are compared. Results: The orientation of the vessel system indicates a homogenous vessel orientation for AB and FB. The average mineral distance (50%) to the closest vessel boundary is higher in AB than in FB (the mean is 96 μm for AB vs. 76 μm for FB; p = 0.021). Average osteocyte lacunar density is found to be higher in AB than in FB (mean 22,874 mm3 vs. 19,376 mm3 for FB; p = 0.038), which might compensate for the high distance from the mineral to the nearest vessel. No significant differences in lacunar volume are found between paired AB and FB. Discussion: A comparable vessel network and similar distribution of vessel porosity between AB and FB may allow the FB graft to exhibit a high regeneration potential when connected to AB, and this might correlate with a high osteoinductive and osteoconductive potential of FB when connected to AB. Since widely used and potent synthetic bone grafts exist, new insight into the bone structure of well-established autologous bone grafts, such as the free fibula flap, could help to improve the performance of such materials and therefore the design of 3D scaffolds.
Collapse
Affiliation(s)
- Jonas Wüster
- Department of Oral and Maxillofacial Surgery, Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Bernhard Hesse
- Xploraytion GmbH, Berlin, Germany
- European Synchrotron Radiation Facility, Grenoble, France
| | - Rene Rothweiler
- Department of Oral- and Craniomaxillofacial Surgery, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | | | - Christian Gross
- Department of Oral- and Craniomaxillofacial Surgery, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | | | | | - Elodie Boller
- European Synchrotron Radiation Facility, Grenoble, France
| | | | - Carsten Rendenbach
- Department of Oral and Maxillofacial Surgery, Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Tobias Fretwurst
- Department of Oral- and Craniomaxillofacial Surgery, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Saskia Preissner
- Department of Oral and Maxillofacial Surgery, Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Max Heiland
- Department of Oral and Maxillofacial Surgery, Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Katja Nelson
- Department of Oral- and Craniomaxillofacial Surgery, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Susanne Nahles
- Department of Oral and Maxillofacial Surgery, Berlin Institute of Health, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
32
|
Hegazy MA, Khairy HM, Hegazy AA, Sebaei MAEF, Sadek SI. Talus bone: normal anatomy, anatomical variations and clinical correlations. Anat Sci Int 2023; 98:391-406. [PMID: 37017903 DOI: 10.1007/s12565-023-00712-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/28/2023] [Indexed: 04/06/2023]
Abstract
Talus is a pivotal bone that assists in easy and correct locomotion and transfers body weight from the shin to the foot. Despite its small size, it is implicated in many clinical disorders. Familiarity with the anatomy of the talus and its anatomical variations is essential for the diagnosis of any disorder related to these variations. Furthermore, orthopedic surgeons must be fully aware by this anatomy during podiatry procedures. In this review, we attempt to present its anatomy in a simple, updated and comprehensive manner. We have also added the anatomical variations and some clinical points relevant to the unique and complex anatomy of talus. The talus has no muscle attachment. However, it does have many ligaments attached to it and others around it to keep it in place. Moreover, the bone plays a pig role in movements due to its involvement in many joints. Most of its surface is covered with articular cartilage. Therefore, its blood supply is relatively poor. This puts the talus at greater risk for poor healing as well as more complications in the event of injury than any other bone. We hope this review will make it easier for clinicians to pursue and understand the updated essential knowledge of one of the most complex bone anatomies that they need in their clinical practice.
Collapse
Affiliation(s)
| | - Hossam Mohammed Khairy
- Orthopedic Surgery Department, Faculty of Medicine, Zagazig University, Zagazig City, 44519, Egypt
| | - Abdelmonem Awad Hegazy
- Basic Medical Science Department, Faculty of Dentistry, Zarqa University, Zarqa City, 13110, Jordan.
- Human Anatomy and Embryology Department, Faculty of Medicine, Zagazig University, Zagazig City, 44519, Egypt.
| | | | - Sami Ibrahim Sadek
- Orthopedic Surgery Department, Faculty of Medicine, Zagazig University, Zagazig City, 44519, Egypt
| |
Collapse
|
33
|
Xu Z, Kusumbe AP, Cai H, Wan Q, Chen J. Type H blood vessels in coupling angiogenesis-osteogenesis and its application in bone tissue engineering. J Biomed Mater Res B Appl Biomater 2023; 111:1434-1446. [PMID: 36880538 DOI: 10.1002/jbm.b.35243] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 02/13/2023] [Accepted: 02/23/2023] [Indexed: 03/08/2023]
Abstract
One specific capillary subtype, termed type H vessel, has been found with unique functional characteristics in coupling angiogenesis with osteogenesis. Researchers have fabricated a variety of tissue engineering scaffolds to enhance bone healing and regeneration through the accumulation of type H vessels. However, only a limited number of reviews discussed the tissue engineering strategies for type H vessel regulation. The object of this review is to summary the current utilizes of bone tissue engineering to regulate type H vessels through various signal pathways including Notch, PDGF-BB, Slit3, HIF-1α, and VEGF signaling. Moreover, we give an insightful overview of recent research progress about the morphological, spatial and age-dependent characteristics of type H blood vessels. Their unique role in tying angiogenesis and osteogenesis together via blood flow, cellular microenvironment, immune system and nervous system are also summarized. This review article would provide an insight into the combination of tissue engineering scaffolds with type H vessels and identify future perspectives for vasculized tissue engineering research.
Collapse
Affiliation(s)
- Zhengyi Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- West China School of Stomatology, Sichuan University, Chengdu, China
| | - Anjali P Kusumbe
- Medical Research Council (MRC) Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), University of Oxford, Oxford, UK
| | - He Cai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- West China School of Stomatology, Sichuan University, Chengdu, China
| | - Qianbing Wan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- West China School of Stomatology, Sichuan University, Chengdu, China
| | - Junyu Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- West China School of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
34
|
Wu X, Zhai F, Chang A, Wei J, Guo Y, Zhang J. Development of Machine Learning Models for Predicting Osteoporosis in Patients with Type 2 Diabetes Mellitus-A Preliminary Study. Diabetes Metab Syndr Obes 2023; 16:1987-2003. [PMID: 37408729 PMCID: PMC10319347 DOI: 10.2147/dmso.s406695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 06/22/2023] [Indexed: 07/07/2023] Open
Abstract
Purpose Diagnosing osteoporosis in T2DM based on bone mineral density (BMD) remains challenging. We sought to develop prediction models employing machine learning algorithms for use as screening instruments for osteoporosis in T2DM patients. Patients and Methods Data were collected from 433 participants and analyzed using nine categorical machine learning algorithms to select features based on demographic and clinical variables. Multiple classification models were compared using the area under the receiver operating characteristic curve (ROC-AUC), accuracy, sensitivity, specificity, the average precision (AP), precision, F1 score, precision-recall curves, calibration plots, and decision curve analysis (DCA) to determine the best model. In addition, 5-fold cross-validation was utilized to optimize the model, followed by an evaluation of feature significance using Shapley Additive exPlanations (SHAP). Using latent class analysis (LCA), distinct subpopulations were identified by constructing several discrete clusters. Results In this study, nine feature variables were identified to construct predictive models for osteoporosis in individuals with T2DM. The machine learning algorithms achieved an AP range of 0.444-1.000. The XGBoost model was selected as the final prediction model with an AUROC of 0.940 in the training set, 0.772 in the validation set for 5-fold cross-validation, and 0.872 in the test set. Using SHAP methodology, 25(OH)D was identified as the most important risk factor. Additionally, a 3-Class model was constructed using LCA, which categorized individuals into high, medium, and low-risk groups. Conclusion Our study developed a predictive model with high accuracy and clinical validity for predicting osteoporosis in type 2 diabetes patients. We also identified three subpopulations with varying osteoporosis risk using clustering. However, limited sample size warrants cautious interpretation of results, and validation in larger cohorts is needed.
Collapse
Affiliation(s)
- Xuelun Wu
- Department of Endocrinology, Cangzhou Central Hospital, Cangzhou City, Hebei Province, People’s Republic of China
| | - Furui Zhai
- Gynecological Clinic, Cangzhou Central Hospital, Cangzhou City, Hebei Province, People’s Republic of China
| | - Ailing Chang
- Department of Endocrinology, Cangzhou Central Hospital, Cangzhou City, Hebei Province, People’s Republic of China
| | - Jing Wei
- Department of Endocrinology, Cangzhou Central Hospital, Cangzhou City, Hebei Province, People’s Republic of China
| | - Yanan Guo
- Department of Endocrinology, Cangzhou Central Hospital, Cangzhou City, Hebei Province, People’s Republic of China
| | - Jincheng Zhang
- Department of Endocrinology, Cangzhou Central Hospital, Cangzhou City, Hebei Province, People’s Republic of China
| |
Collapse
|
35
|
Zhang H, Wu S, Chen W, Hu Y, Geng Z, Su J. Bone/cartilage targeted hydrogel: Strategies and applications. Bioact Mater 2023; 23:156-169. [PMID: 36406248 PMCID: PMC9661677 DOI: 10.1016/j.bioactmat.2022.10.028] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/13/2022] Open
Abstract
The skeletal system is responsible for weight-bearing, organ protection, and movement. Bone diseases caused by trauma, infection, and aging can seriously affect a patient's quality of life. Bone targeted biomaterials are suitable for the treatment of bone diseases. Biomaterials with bone-targeted properties can improve drug utilization and reduce side effects. A large number of bone-targeted micro-nano materials have been developed. However, only a few studies addressed bone-targeted hydrogel. The large size of hydrogel makes it difficult to achieve systematic targeting. However, local targeted hydrogel still has significant prospects. Molecules in bone/cartilage extracellular matrix and bone cells provide binding sites for bone-targeted hydrogel. Drug delivery systems featuring microgels with targeting properties is a key construction strategy for bone-targeted hydrogel. Besides, injectable hydrogel drug depot carrying bone-targeted drugs is another strategy. In this review, we summarize the bone-targeted hydrogel through application environment, construction strategies and disease applications. We hope this article will provide a reference for the development of bone-targeted hydrogels. We also hope this article could increase awareness of bone-targeted materials. Introducing the microenvironment and target molecules in different parts of long bones. Summarizing the construction strategy of micro/nanoparticle hydrogel with bone targeting properties. Summarizing the construction strategy of hydrogel based depot carrying bone-targeted drugs. Reporting the application and effect of bone targeting hydrogel in common bone diseases.
Collapse
|
36
|
Ni N, Ge M, Huang R, Zhang D, Lin H, Ju Y, Tang Z, Gao H, Zhou H, Chen Y, Gu P. Thermodynamic 2D Silicene for Sequential and Multistage Bone Regeneration. Adv Healthc Mater 2023; 12. [DOI: doi.org/10.1002/adhm.202203107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Indexed: 09/08/2023]
Abstract
AbstractBone healing is a multistage process involving the recruitment of cells, revascularization, and osteogenic differentiation, all of which are modulated in the temporal sequence to maximize cascade bone regeneration. However, insufficient osteoblast cells, poor blood supply, and limited bone induction at the site of critical‐sized bone defect broadly impede bone repair. 2D SiO2‐silicene@2,2′‐,azobis(2‐[2‐imidazolin‐2‐yl] propane) (SNSs@AIPH) with inherent thermodynamic property and osteoinductive activity is therefore designed and engineered for sequentially efficient bone repair. By means of controllable NIR‐II irradiation, the integrated SNSs@AIPH stimulates the generation of appropriate intracellular reactive oxygen species, which accelerates early bone marrow mesenchymal stem cells (BMSCs) proliferation and angiogenesis remarkably. Importantly, as silicon‐based 2D nanoparticles, the engineered SNSs@AIPH with high biocompatibility features distinct bioactivity to significantly promote BMSCs osteogenesis differentiation by activating TGFβ and BMP pathways. In a rat cranial defect model, SNSs@AIPH‐NIR‐II leads to a comparable increase of BMSCs proliferation and local vascularization at an early stage, followed by significant osteogenic differentiation, synergically resulting in a highly effective bone repair. Collectively, the fascinating characteristics and exceptional bone repair efficiency of NIR‐II‐mediated SNSs@AIPH allow it to be a promising bionic‐oriented strategy for bone regeneration, broadening a new perspective in the application of cell‐instructive biomaterials in bone tissue engineering.
Collapse
Affiliation(s)
- Ni Ni
- Department of Ophthalmology Shanghai Ninth People's Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200011 P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology Shanghai 200011 P. R. China
| | - Min Ge
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Rui Huang
- Department of Ophthalmology Shanghai Ninth People's Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200011 P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology Shanghai 200011 P. R. China
| | - Dandan Zhang
- Department of Ophthalmology Shanghai Ninth People's Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200011 P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology Shanghai 200011 P. R. China
| | - Han Lin
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
| | - Yahan Ju
- Department of Ophthalmology Shanghai Ninth People's Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200011 P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology Shanghai 200011 P. R. China
| | - Zhimin Tang
- Department of Ophthalmology Shanghai Ninth People's Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200011 P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology Shanghai 200011 P. R. China
| | - Huiqin Gao
- Department of Ophthalmology Shanghai Ninth People's Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200011 P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology Shanghai 200011 P. R. China
| | - Huifang Zhou
- Department of Ophthalmology Shanghai Ninth People's Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200011 P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology Shanghai 200011 P. R. China
| | - Yu Chen
- Materdicine Lab School of Life Sciences Shanghai University Shanghai 200444 P. R. China
| | - Ping Gu
- Department of Ophthalmology Shanghai Ninth People's Hospital Shanghai Jiao Tong University School of Medicine Shanghai 200011 P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology Shanghai 200011 P. R. China
| |
Collapse
|
37
|
Ni N, Ge M, Huang R, Zhang D, Lin H, Ju Y, Tang Z, Gao H, Zhou H, Chen Y, Gu P. Thermodynamic 2D Silicene for Sequential and Multistage Bone Regeneration. Adv Healthc Mater 2023; 12:e2203107. [PMID: 36690338 PMCID: PMC11468546 DOI: 10.1002/adhm.202203107] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/14/2023] [Indexed: 01/25/2023]
Abstract
Bone healing is a multistage process involving the recruitment of cells, revascularization, and osteogenic differentiation, all of which are modulated in the temporal sequence to maximize cascade bone regeneration. However, insufficient osteoblast cells, poor blood supply, and limited bone induction at the site of critical-sized bone defect broadly impede bone repair. 2D SiO2 -silicene@2,2'-,azobis(2-[2-imidazolin-2-yl] propane) (SNSs@AIPH) with inherent thermodynamic property and osteoinductive activity is therefore designed and engineered for sequentially efficient bone repair. By means of controllable NIR-II irradiation, the integrated SNSs@AIPH stimulates the generation of appropriate intracellular reactive oxygen species, which accelerates early bone marrow mesenchymal stem cells (BMSCs) proliferation and angiogenesis remarkably. Importantly, as silicon-based 2D nanoparticles, the engineered SNSs@AIPH with high biocompatibility features distinct bioactivity to significantly promote BMSCs osteogenesis differentiation by activating TGFβ and BMP pathways. In a rat cranial defect model, SNSs@AIPH-NIR-II leads to a comparable increase of BMSCs proliferation and local vascularization at an early stage, followed by significant osteogenic differentiation, synergically resulting in a highly effective bone repair. Collectively, the fascinating characteristics and exceptional bone repair efficiency of NIR-II-mediated SNSs@AIPH allow it to be a promising bionic-oriented strategy for bone regeneration, broadening a new perspective in the application of cell-instructive biomaterials in bone tissue engineering.
Collapse
Affiliation(s)
- Ni Ni
- Department of OphthalmologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghai200011P. R. China
| | - Min Ge
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institute of Ceramics Chinese Academy of SciencesShanghai200050P. R. China
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijing100049P. R. China
| | - Rui Huang
- Department of OphthalmologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghai200011P. R. China
| | - Dandan Zhang
- Department of OphthalmologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghai200011P. R. China
| | - Han Lin
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institute of Ceramics Chinese Academy of SciencesShanghai200050P. R. China
| | - Yahan Ju
- Department of OphthalmologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghai200011P. R. China
| | - Zhimin Tang
- Department of OphthalmologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghai200011P. R. China
| | - Huiqin Gao
- Department of OphthalmologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghai200011P. R. China
| | - Huifang Zhou
- Department of OphthalmologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghai200011P. R. China
| | - Yu Chen
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
| | - Ping Gu
- Department of OphthalmologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghai200011P. R. China
| |
Collapse
|
38
|
Yusoff NA, Abd Hamid Z, Budin SB, Taib IS. Linking Benzene, in Utero Carcinogenicity and Fetal Hematopoietic Stem Cell Niches: A Mechanistic Review. Int J Mol Sci 2023; 24:ijms24076335. [PMID: 37047305 PMCID: PMC10094243 DOI: 10.3390/ijms24076335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/19/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
Previous research reported that prolonged benzene exposure during in utero fetal development causes greater fetal abnormalities than in adult-stage exposure. This phenomenon increases the risk for disease development at the fetal stage, particularly carcinogenesis, which is mainly associated with hematological malignancies. Benzene has been reported to potentially act via multiple modes of action that target the hematopoietic stem cell (HSCs) niche, a complex microenvironment in which HSCs and multilineage hematopoietic stem and progenitor cells (HSPCs) reside. Oxidative stress, chromosomal aberration and epigenetic modification are among the known mechanisms mediating benzene-induced genetic and epigenetic modification in fetal stem cells leading to in utero carcinogenesis. Hence, it is crucial to monitor exposure to carcinogenic benzene via environmental, occupational or lifestyle factors among pregnant women. Benzene is a well-known cause of adult leukemia. However, proof of benzene involvement with childhood leukemia remains scarce despite previously reported research linking incidences of hematological disorders and maternal benzene exposure. Furthermore, accumulating evidence has shown that maternal benzene exposure is able to alter the developmental and functional properties of HSPCs, leading to hematological disorders in fetus and children. Since HSPCs are parental blood cells that regulate hematopoiesis during the fetal and adult stages, benzene exposure that targets HSPCs may induce damage to the population and trigger the development of hematological diseases. Therefore, the mechanism of in utero carcinogenicity by benzene in targeting fetal HSPCs is the primary focus of this review.
Collapse
|
39
|
Zhang H, Liesveld JL, Calvi LM, Lipe BC, Xing L, Becker MW, Schwarz EM, Yeh SCA. The roles of bone remodeling in normal hematopoiesis and age-related hematological malignancies. Bone Res 2023; 11:15. [PMID: 36918531 PMCID: PMC10014945 DOI: 10.1038/s41413-023-00249-w] [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: 08/01/2022] [Revised: 12/24/2022] [Accepted: 01/26/2023] [Indexed: 03/16/2023] Open
Abstract
Prior research establishing that bone interacts in coordination with the bone marrow microenvironment (BMME) to regulate hematopoietic homeostasis was largely based on analyses of individual bone-associated cell populations. Recent advances in intravital imaging has suggested that the expansion of hematopoietic stem cells (HSCs) and acute myeloid leukemia cells is restricted to bone marrow microdomains during a distinct stage of bone remodeling. These findings indicate that dynamic bone remodeling likely imposes additional heterogeneity within the BMME to yield differential clonal responses. A holistic understanding of the role of bone remodeling in regulating the stem cell niche and how these interactions are altered in age-related hematological malignancies will be critical to the development of novel interventions. To advance this understanding, herein, we provide a synopsis of the cellular and molecular constituents that participate in bone turnover and their known connections to the hematopoietic compartment. Specifically, we elaborate on the coupling between bone remodeling and the BMME in homeostasis and age-related hematological malignancies and after treatment with bone-targeting approaches. We then discuss unresolved questions and ambiguities that remain in the field.
Collapse
Affiliation(s)
- Hengwei Zhang
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA.
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA.
| | - Jane L Liesveld
- Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Hematology/Oncology and Bone Marrow Transplantation Program, University of Rochester Medical Center, Rochester, NY, USA
| | - Laura M Calvi
- Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Endocrinology/Metabolism, University of Rochester Medical Center, Rochester, NY, USA
| | - Brea C Lipe
- Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Hematology/Oncology and Bone Marrow Transplantation Program, University of Rochester Medical Center, Rochester, NY, USA
| | - Lianping Xing
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Michael W Becker
- Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Hematology/Oncology and Bone Marrow Transplantation Program, University of Rochester Medical Center, Rochester, NY, USA
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, Division of Allergy/Immunology/Rheumatology, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Shu-Chi A Yeh
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Box 665, Rochester, NY, 14642, USA.
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA.
- Department of Physiology/Pharmacology, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
40
|
Zhao YC, Zhang Y, Jiang F, Wu C, Wan B, Syeda R, Li Q, Shen B, Ju LA. A Novel Computational Biomechanics Framework to Model Vascular Mechanopropagation in Deep Bone Marrow. Adv Healthc Mater 2023; 12:e2201830. [PMID: 36521080 PMCID: PMC11469229 DOI: 10.1002/adhm.202201830] [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: 08/18/2022] [Revised: 12/05/2022] [Indexed: 12/23/2022]
Abstract
The mechanical stimuli generated by body exercise can be transmitted from cortical bone into the deep bone marrow (mechanopropagation). Excitingly, a mechanosensitive perivascular stem cell niche is recently identified within the bone marrow for osteogenesis and lymphopoiesis. Although it is long known that they are maintained by exercise-induced mechanical stimulation, the mechanopropagation from compact bone to deep bone marrow vasculature remains elusive of this fundamental mechanobiology field. No experimental system is available yet to directly understand such exercise-induced mechanopropagation at the bone-vessel interface. To this end, taking advantage of the revolutionary in vivo 3D deep bone imaging, an integrated computational biomechanics framework to quantitatively evaluate the mechanopropagation capabilities for bone marrow arterioles, arteries, and sinusoids is devised. As a highlight, the 3D geometries of blood vessels are smoothly reconstructed in the presence of vessel wall thickness and intravascular pulse pressure. By implementing the 5-parameter Mooney-Rivlin model that simulates the hyperelastic vessel properties, finite element analysis to thoroughly investigate the mechanical effects of exercise-induced intravascular vibratory stretching on bone marrow vasculature is performed. In addition, the blood pressure and cortical bone bending effects on vascular mechanoproperties are examined. For the first time, movement-induced mechanopropagation from the hard cortical bone to the soft vasculature in the bone marrow is numerically simulated. It is concluded that arterioles and arteries are much more efficient in propagating mechanical force than sinusoids due to their stiffness. In the future, this in-silico approach can be combined with other clinical imaging modalities for subject/patient-specific vascular reconstruction and biomechanical analysis, providing large-scale phenotypic data for personalized mechanobiology discovery.
Collapse
Affiliation(s)
- Yunduo Charles Zhao
- School of Biomedical EngineeringThe University of Sydney2008New South WalesDarlingtonAustralia
- Charles Perkins CentreThe University of Sydney2006New South WalesCamperdownAustralia
- The University of Sydney Nano Institute (Sydney Nano)The University of Sydney2006New South WalesCamperdownAustralia
| | - Yingqi Zhang
- School of Biomedical EngineeringThe University of Sydney2008New South WalesDarlingtonAustralia
- Charles Perkins CentreThe University of Sydney2006New South WalesCamperdownAustralia
- The University of Sydney Nano Institute (Sydney Nano)The University of Sydney2006New South WalesCamperdownAustralia
| | - Fengtao Jiang
- School of Biomedical EngineeringThe University of Sydney2008New South WalesDarlingtonAustralia
- Charles Perkins CentreThe University of Sydney2006New South WalesCamperdownAustralia
- The University of Sydney Nano Institute (Sydney Nano)The University of Sydney2006New South WalesCamperdownAustralia
| | - Chi Wu
- School of Aerospace, Mechanical and Mechatronic EngineeringThe University of Sydney2008New South WalesDarlingtonAustralia
| | - Boyang Wan
- School of Aerospace, Mechanical and Mechatronic EngineeringThe University of Sydney2008New South WalesDarlingtonAustralia
| | - Ruhma Syeda
- Department of NeuroscienceUniversity of Texas Southwestern Medical Center75235TXDallasUSA
| | - Qing Li
- School of Aerospace, Mechanical and Mechatronic EngineeringThe University of Sydney2008New South WalesDarlingtonAustralia
| | - Bo Shen
- National Institute of Biological ScienceZhongguancun Life Science Park102206BeijingChina
- Tsinghua Institute of Multidisciplinary Biomedical ResearchTsinghua University102206BeijingChina
| | - Lining Arnold Ju
- School of Biomedical EngineeringThe University of Sydney2008New South WalesDarlingtonAustralia
- Charles Perkins CentreThe University of Sydney2006New South WalesCamperdownAustralia
- The University of Sydney Nano Institute (Sydney Nano)The University of Sydney2006New South WalesCamperdownAustralia
| |
Collapse
|
41
|
Gerami MH, Khorram R, Rasoolzadegan S, Mardpour S, Nakhaei P, Hashemi S, Al-Naqeeb BZT, Aminian A, Samimi S. Emerging role of mesenchymal stem/stromal cells (MSCs) and MSCs-derived exosomes in bone- and joint-associated musculoskeletal disorders: a new frontier. Eur J Med Res 2023; 28:86. [PMID: 36803566 PMCID: PMC9939872 DOI: 10.1186/s40001-023-01034-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 01/26/2023] [Indexed: 02/22/2023] Open
Abstract
Exosomes are membranous vesicles with a 30 to 150 nm diameter secreted by mesenchymal stem/stromal cells (MSCs) and other cells, such as immune cells and cancer cells. Exosomes convey proteins, bioactive lipids, and genetic components to recipient cells, such as microRNAs (miRNAs). Consequently, they have been implicated in regulating intercellular communication mediators under physiological and pathological circumstances. Exosomes therapy as a cell-free approach bypasses many concerns regarding the therapeutic application of stem/stromal cells, including undesirable proliferation, heterogeneity, and immunogenic effects. Indeed, exosomes have become a promising strategy to treat human diseases, particularly bone- and joint-associated musculoskeletal disorders, because of their characteristics, such as potentiated stability in circulation, biocompatibility, low immunogenicity, and toxicity. In this light, a diversity of studies have indicated that inhibiting inflammation, inducing angiogenesis, provoking osteoblast and chondrocyte proliferation and migration, and negative regulation of matrix-degrading enzymes result in bone and cartilage recovery upon administration of MSCs-derived exosomes. Notwithstanding, insufficient quantity of isolated exosomes, lack of reliable potency test, and exosomes heterogeneity hurdle their application in clinics. Herein, we will deliver an outline respecting the advantages of MSCs-derived exosomes-based therapy in common bone- and joint-associated musculoskeletal disorders. Moreover, we will have a glimpse the underlying mechanism behind the MSCs-elicited therapeutic merits in these conditions.
Collapse
Affiliation(s)
- Mohammad Hadi Gerami
- grid.412571.40000 0000 8819 4698Bone and Joint Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Roya Khorram
- grid.412571.40000 0000 8819 4698Bone and Joint Diseases Research Center, Department of Orthopedic Surgery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soheil Rasoolzadegan
- grid.411600.2Department of Surgery, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeid Mardpour
- grid.411705.60000 0001 0166 0922Department of Radiology, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Pooria Nakhaei
- grid.411705.60000 0001 0166 0922Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Soheyla Hashemi
- grid.411036.10000 0001 1498 685XObstetrician, Gynaecology & Infertility Department, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Amir Aminian
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences, Tehran, Iran.
| | - Sahar Samimi
- Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
42
|
Biswas L, Chen J, De Angelis J, Singh A, Owen-Woods C, Ding Z, Pujol JM, Kumar N, Zeng F, Ramasamy SK, Kusumbe AP. Lymphatic vessels in bone support regeneration after injury. Cell 2023; 186:382-397.e24. [PMID: 36669473 PMCID: PMC11913777 DOI: 10.1016/j.cell.2022.12.031] [Citation(s) in RCA: 101] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 10/05/2022] [Accepted: 12/19/2022] [Indexed: 01/20/2023]
Abstract
Blood and lymphatic vessels form a versatile transport network and provide inductive signals to regulate tissue-specific functions. Blood vessels in bone regulate osteogenesis and hematopoiesis, but current dogma suggests that bone lacks lymphatic vessels. Here, by combining high-resolution light-sheet imaging and cell-specific mouse genetics, we demonstrate presence of lymphatic vessels in mouse and human bones. We find that lymphatic vessels in bone expand during genotoxic stress. VEGF-C/VEGFR-3 signaling and genotoxic stress-induced IL6 drive lymphangiogenesis in bones. During lymphangiogenesis, secretion of CXCL12 from proliferating lymphatic endothelial cells is critical for hematopoietic and bone regeneration. Moreover, lymphangiocrine CXCL12 triggers expansion of mature Myh11+ CXCR4+ pericytes, which differentiate into bone cells and contribute to bone and hematopoietic regeneration. In aged animals, such expansion of lymphatic vessels and Myh11-positive cells in response to genotoxic stress is impaired. These data suggest lymphangiogenesis as a therapeutic avenue to stimulate hematopoietic and bone regeneration.
Collapse
Affiliation(s)
- Lincoln Biswas
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK
| | - Junyu Chen
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jessica De Angelis
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK
| | - Amit Singh
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK; Heidelberg University Biochemistry Center, Im Neuenheimer Feld 328, Heidelberg D-69120, Germany
| | - Charlotte Owen-Woods
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK
| | - Zhangfan Ding
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Joan Mane Pujol
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK
| | - Naveen Kumar
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK
| | - Fanxin Zeng
- Department of Clinic Medical Center, Dazhou Central Hospital, Dazhou, China
| | - Saravana K Ramasamy
- MRC London Institute of Medical Sciences, Imperial College London, London W12 0NN, UK
| | - Anjali P Kusumbe
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Medical Sciences Division, University of Oxford, Oxford OX3 9DS, UK.
| |
Collapse
|
43
|
Uysal A, Oktay G, Ural C, Kalkan NB. The effect of ferritin levels on distal femoral cartilage thickness in patients with beta thalassaemia major. J Bone Miner Metab 2023; 41:95-104. [PMID: 36422676 DOI: 10.1007/s00774-022-01384-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 10/21/2022] [Indexed: 11/27/2022]
Abstract
INTRODUCTION To the best of our knowledge, the present study is the first in the literature to assess distal femoral cartilage thickness and its relationship with ferritin levels in adult patients with beta thalassaemia major (BTM). MATERIALS AND METHODS 45 patients with BTM and 45 healthy controls were included in the study. Ferritin and haemoglobin levels of the patient and healthy groups were determined by blood analysis and distal femoral cartilage thicknesses were measured via ultrasound. Then, the patient group was divided into subgroups according to whether their ferritin levels were below or above 2500 µg/L. They were then compared among themselves and with the healthy control group using the available data. RESULTS Distal femoral cartilage thickness values were statistically significantly lower in the BTM group compared to the healthy control group (p values < 0.001). Patients with a ferritin level below 2500 µg/L had statistically significantly higher right and left average distal femoral cartilage thickness values than the patients with a ferritin level above 2500 µg/L (p = 0.029 and p = 0.019, respectively). The right and left average distal femoral cartilage thickness values of the patient subgroup with low ferritin levels were statistically similar to the control group (p = 0.146 and p = 0.164, respectively). CONCLUSION Our study showed that thalassaemia patients are more likely to develop osteoarthritis (OA) than the normal population and possible OA development can be prevented by keeping the ferritin levels of these patients in the optimum range.
Collapse
Affiliation(s)
- Alper Uysal
- Physical Medicine and Rehabilitation Clinic, Hatay Training and Research Hospital, Güzelburç, 31001, Antakya, Hatay, Turkey.
| | - Gönül Oktay
- Thalassemia Center, Hatay Training and Research Hospital, Hatay, Turkey
| | - Cihan Ural
- Hematology Clinic, Hatay Training and Research Hospital, Hatay, Turkey
| | | |
Collapse
|
44
|
Montevecchi M, Marucci G, Pignataro B, Piana G, Alessandri-Bonetti G, Checchi V. Bone Modeling after Orthodontic Extrusion: A Histomorphometric Pilot Study. J Clin Med 2022; 11:jcm11247329. [PMID: 36555945 PMCID: PMC9781502 DOI: 10.3390/jcm11247329] [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: 09/23/2022] [Revised: 12/03/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
During osteogenesis and bone modeling, high vascularity and osteoblastic/osteoclastic cell activity have been detected. A decrease in this activity is a sign of complete bone formation and maturation. Alveolar bone maturation seems to occur within weeks and months; however, the precise timing of the alveolar bone modeling is still unknown. The aim of this clinical pilot study was to investigate the bone modeling of neo-apposed tissue during orthodontic extrusive movements, through a histomorphometric analysis of human biopsies. This study was conducted on third mandibular molars sockets, and all teeth were extracted after orthodontic extrusion between 2010 and 2014. After different stabilization timings, extractions were performed, and a specimen of neo-deposed bone was harvested from each socket for the histomorphometric analysis. Histological parameters were evaluated to identify bone quantity and quality. This study included 12 teeth extracted from 9 patients. All specimens were composed of bone tissue. Bone samples taken after 1 and 1.5 months of stabilization presented remarkable percentages of woven bone, while after 2 months, a relevant decrease was observed. Histomorphometric analysis suggested that after orthodontic extrusion, a period of stabilization of 2 months allows the neo-deposed bone to mature.
Collapse
Affiliation(s)
- Marco Montevecchi
- Department of Biomedical and Neuromotor Sciences—DIBINEM, University of Bologna, Via San Vitale 59, 40100 Bologna, Italy
- Correspondence:
| | - Gianluca Marucci
- Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | | | - Gabriela Piana
- Department of Biomedical and Neuromotor Sciences—DIBINEM, University of Bologna, Via San Vitale 59, 40100 Bologna, Italy
| | - Giulio Alessandri-Bonetti
- Department of Biomedical and Neuromotor Sciences—DIBINEM, University of Bologna, Via San Vitale 59, 40100 Bologna, Italy
| | - Vittorio Checchi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, Unit of Dentistry and Oral-Maxillo-Facial Surgery, University of Modena and Reggio Emilia, Via del Pozzo 71, 41100 Modena, Italy
| |
Collapse
|
45
|
An S, Zheng S, Cai Z, Chen S, Wang C, Li Y, Deng Z. Connexin43 in Musculoskeletal System: New Targets for Development and Disease Progression. Aging Dis 2022; 13:1715-1732. [PMID: 36465186 PMCID: PMC9662276 DOI: 10.14336/ad.2022.0421] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/21/2022] [Indexed: 03/16/2025] Open
Abstract
Connexin43, which is the most highly expressed connexin subtype in the musculoskeletal system, exists in a variety of bone cells, synovial tissue, and cartilage tissue. Connexin43 has been suggested to be a key regulator of bone homeostasis. Studies have shown aberrant Connexin43 expression in musculoskeletal disorders, such as osteoporosis, osteoarthritis, and rheumatoid arthritis. During cellular activities, Connexin43 can participate in the formation of functionally specific gap junctions and hemichannels and can exert independent cellular regulatory and signaling functions through special C-termini. The critical role of Connexin43 in physiological development and disease progression has been gradually revealed. In this article, the function of Connexin43 in musculoskeletal tissues is summarized, revealing the potential role of Connexin43 as a key target in the treatment of related bone and muscle disorders and the need for further discovery.
Collapse
Affiliation(s)
- Senbo An
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| | - Shengyuan Zheng
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha, Hunan, China.
| | - Zijun Cai
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Siyu Chen
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China.
| | - Chen Wang
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China.
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Zhenhan Deng
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China.
| |
Collapse
|
46
|
Faienza MF, Pontrelli P, Brunetti G. Type 2 diabetes and bone fragility in children and adults. World J Diabetes 2022; 13:900-911. [PMID: 36437868 PMCID: PMC9693736 DOI: 10.4239/wjd.v13.i11.900] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/17/2022] [Accepted: 10/11/2022] [Indexed: 11/11/2022] Open
Abstract
Type 2 diabetes (T2D) is a global epidemic disease. The prevalence of T2D in adolescents and young adults is increasing alarmingly. The mechanisms leading to T2D in young people are similar to those in older patients. However, the severity of onset, reduced insulin sensitivity and defective insulin secretion can be different in subjects who develop the disease at a younger age. T2D is associated with different complications, including bone fragility with consequent susceptibility to fractures. The purpose of this systematic review was to describe T2D bone fragility together with all the possible involved pathways. Numerous studies have reported that patients with T2D show preserved, or even increased, bone mineral density compared with controls. This apparent paradox can be explained by the altered bone quality with increased cortical bone porosity and compr-omised mechanical properties. Furthermore, reduced bone turnover has been described in T2D with reduced markers of bone formation and resorption. These findings prompted different researchers to highlight the mechanisms leading to bone fragility, and numerous critical altered pathways have been identified and studied. In detail, we focused our attention on the role of microvascular disease, advanced glycation end products, the senescence pathway, the Wnt/β-catenin pathway, the osteoprotegerin/receptor-activator of nuclear factor kappa B ligand, osteonectin and fibroblast growth factor 23. The understanding of type 2 myeloid bone fragility is an important issue as it could suggest possible interventions for the prevention of poor bone quality in T2D and/or how to target these pathways when bone disease is clearly evident.
Collapse
Affiliation(s)
- Maria Felicia Faienza
- Department of Biomedical Sciences and Human Oncology, Pediatric Unit, University of Bari Aldo Moro, Bari 70124, Italy
| | - Paola Pontrelli
- Division of Nephrology, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari 70124, Italy
| | - Giacomina Brunetti
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari 70125, Italy
| |
Collapse
|
47
|
Schilling K, Zhai Y, Zhou Z, Zhou B, Brown E, Zhang X. High-resolution imaging of the osteogenic and angiogenic interface at the site of murine cranial bone defect repair via multiphoton microscopy. eLife 2022; 11:e83146. [PMID: 36326085 PMCID: PMC9678361 DOI: 10.7554/elife.83146] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/31/2022] [Indexed: 11/05/2022] Open
Abstract
The spatiotemporal blood vessel formation and specification at the osteogenic and angiogenic interface of murine cranial bone defect repair were examined utilizing a high-resolution multiphoton-based imaging platform in conjunction with advanced optical techniques that allow interrogation of the oxygen microenvironment and cellular energy metabolism in living animals. Our study demonstrates the dynamic changes of vessel types, that is, arterial, venous, and capillary vessel networks at the superior and dura periosteum of cranial bone defect, suggesting a differential coupling of the vessel type with osteoblast expansion and bone tissue deposition/remodeling during repair. Employing transgenic reporter mouse models that label distinct types of vessels at the site of repair, we further show that oxygen distributions in capillary vessels at the healing site are heterogeneous as well as time- and location-dependent. The endothelial cells coupling to osteoblasts prefer glycolysis and are less sensitive to microenvironmental oxygen changes than osteoblasts. In comparison, osteoblasts utilize relatively more OxPhos and potentially consume more oxygen at the site of repair. Taken together, our study highlights the dynamics and functional significance of blood vessel types at the site of defect repair, opening up opportunities for further delineating the oxygen and metabolic microenvironment at the interface of bone tissue regeneration.
Collapse
Affiliation(s)
- Kevin Schilling
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and DentistryRochesterUnited States
- Department of Biomedical Engineering, University of RochesterRochesterUnited States
| | - Yuankun Zhai
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and DentistryRochesterUnited States
| | - Zhuang Zhou
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and DentistryRochesterUnited States
| | - Bin Zhou
- Shanghai Institutes for Biological SciencesShanghaiChina
| | - Edward Brown
- Department of Biomedical Engineering, University of RochesterRochesterUnited States
| | - Xinping Zhang
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and DentistryRochesterUnited States
| |
Collapse
|
48
|
Zheng G, Ma HW, Xiang GH, He GL, Cai HC, Dai ZH, Chen YL, Lin Y, Xu HZ, Ni WF, Xu C, Liu HX, Wang XY. Bone-targeting delivery of platelet lysate exosomes ameliorates glucocorticoid-induced osteoporosis by enhancing bone-vessel coupling. J Nanobiotechnology 2022; 20:220. [PMID: 36310171 PMCID: PMC9620632 DOI: 10.1186/s12951-022-01400-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/26/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Glucocorticoids (GCs) overuse is associated with decreased bone mass and osseous vasculature destruction, leading to severe osteoporosis. Platelet lysates (PL) as a pool of growth factors (GFs) were widely used in local bone repair by its potent pro-regeneration and pro-angiogenesis. However, it is still seldom applied for treating systemic osteopathia due to the lack of a suitable delivery strategy. The non-targeted distribution of GFs might cause tumorigenesis in other organs. RESULTS In this study, PL-derived exosomes (PL-exo) were isolated to enrich the platelet-derived GFs, followed by conjugating with alendronate (ALN) grafted PEGylated phospholipid (DSPE-PEG-ALN) to establish a bone-targeting PL-exo (PL-exo-ALN). The in vitro hydroxyapatite binding affinity and in vivo bone targeting aggregation of PL-exo were significantly enhanced after ALN modification. Besides directly modulating the osteogenic and angiogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and endothelial progenitor cells (EPCs), respectively, PL-exo-ALN also facilitate their coupling under GCs' stimulation. Additionally, intravenous injection of PL-exo-ALN could successfully rescue GCs induced osteoporosis (GIOP) in vivo. CONCLUSIONS PL-exo-ALN may be utilized as a novel nanoplatform for precise infusion of GFs to bone sites and exerts promising therapeutic potential for GIOP.
Collapse
Affiliation(s)
- Gang Zheng
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Hai-Wei Ma
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Guang-Heng Xiang
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Gao-Lu He
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Han-Chen Cai
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Zi-Han Dai
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Yan-Lin Chen
- Department of Orthopaedic Surgery, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, Zhejiang Province, China
| | - Yan Lin
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Hua-Zi Xu
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Wen-Fei Ni
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| | - Cong Xu
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| | - Hai-Xiao Liu
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| | - Xiang-Yang Wang
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| |
Collapse
|
49
|
Kim MJ, Valderrábano RJ, Wu JY. Osteoblast Lineage Support of Hematopoiesis in Health and Disease. J Bone Miner Res 2022; 37:1823-1842. [PMID: 35983701 PMCID: PMC11346465 DOI: 10.1002/jbmr.4678] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/21/2022] [Accepted: 08/13/2022] [Indexed: 11/06/2022]
Abstract
In mammals, hematopoiesis migrates to the bone marrow during embryogenesis coincident with the appearance of mineralized bone, where hematopoietic stem cells (HSCs) and their progeny are maintained by the surrounding microenvironment or niche, and sustain the entirety of the hematopoietic system. Genetic manipulation of niche factors and advances in cell lineage tracing techniques have implicated cells of both hematopoietic and nonhematopoietic origin as important regulators of hematopoiesis in health and disease. Among them, cells of the osteoblast lineage, from stromal skeletal stem cells to matrix-embedded osteocytes, are vital niche residents with varying capacities for hematopoietic support depending on stage of differentiation. Here, we review populations of osteoblasts at differing stages of differentiation and summarize the current understanding of the role of the osteoblast lineage in supporting hematopoiesis. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Matthew J Kim
- Division of Endocrinology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Rodrigo J Valderrábano
- Research Program in Men's Health: Aging and Metabolism, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joy Y Wu
- Division of Endocrinology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
50
|
Rodrigues J, Wang YF, Singh A, Hendriks M, Dharmalingam G, Cohen-Solal M, Kusumbe AP, Ramasamy SK. Oestrogen enforces the integrity of blood vessels in the bone during pregnancy and menopause. NATURE CARDIOVASCULAR RESEARCH 2022; 1:918-932. [PMID: 36531334 PMCID: PMC7613952 DOI: 10.1038/s44161-022-00139-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 09/02/2022] [Indexed: 04/27/2025]
Abstract
The mammalian skeletal system shows sex differences in structure, functions, ageing and disease incidences. The role of blood vessels in physiological, regenerative and pathological bone functions indicates the requisite to understanding their sex specificity. Here, we find oestrogen regulates blood vessel physiology during pregnancy and menopause through oestrogen receptor alpha (ERα) and G-protein coupled oestrogen receptor-1 (Gper1) but not ERβ-dependent signalling in mice. Oestrogen regulates BECs' lipid use and promotes lipolysis of adipocytes and FA uptake from the microenvironment. Low oestrogen conditions skew endothelial FA metabolism to accumulate lipid peroxides (LPO), leading to vascular ageing. High ferrous ion levels in female BECs intensify LPO accumulation and accelerate the ageing process. Importantly, inhibiting LPO generation using liproxstatin-1 in aged mice significantly improved bone heath. Thus, our findings illustrate oestrogen's effects on BECs and suggest LPO targeting could be an efficient strategy to manage blood and bone health in females.
Collapse
Affiliation(s)
- Julia Rodrigues
- Institute of Clinical Sciences, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Yi-Fang Wang
- Bioinformatics and Computing Facility, MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Amit Singh
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Heidelberg University Biochemistry Center, University of Heidelberg, Heidelberg, Germany
| | - Michelle Hendriks
- Institute of Clinical Sciences, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Gopuraja Dharmalingam
- Bioinformatics and Computing Facility, MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Martine Cohen-Solal
- Bioscar Inserm U1132 and Université de Paris, Hospital Lariboisiere, Paris, France
| | - Anjali P Kusumbe
- Tissue and Tumor Microenvironments Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Saravana K Ramasamy
- Institute of Clinical Sciences, Imperial College London, London, UK.
- MRC London Institute of Medical Sciences, Imperial College London, London, UK.
| |
Collapse
|