1
|
King JS, Wan M, Kim A, Prabhu S, Novak S, Kalajzic I, Delany AM, Sanjay A. Effects of aging on the immune and periosteal response to fracture injury. Bone 2025; 198:117524. [PMID: 40381878 DOI: 10.1016/j.bone.2025.117524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 04/14/2025] [Accepted: 05/13/2025] [Indexed: 05/20/2025]
Abstract
Aging predisposes individuals to reduced bone mass and fragility fractures, which are costly and linked to high mortality. Understanding how aging affects fracture healing is essential for developing therapies to enhance bone regeneration in older adults. During the inflammatory phase of fracture healing, immune cells are recruited to the injury site as periosteal skeletal stem/progenitor cells (pSSPCs) rapidly proliferate and differentiate into osteochondral lineages, allowing for fibrocartilaginous callus formation and, subsequently, complete bone healing. Irrespective of age, how periosteal mesenchymal and immune cells interact during early fracture healing is incompletely understood, limiting our ability to modulate this process. To address this, we directly analyzed, in parallel, at a single-cell level, isolated murine CD45(+) and CD45(-) periosteal cells dissected from intact and fractured bones, collected three days after injury. Comprehensive analysis, corroborated by bulk RNA-sequencing, flow cytometry, and histology, demonstrated that aging decreased pSSPC proliferation, markedly reduced expression of genes required for callus formation, and increased senescence signature. During the regeneration phase, at 14 days post injury, aged mice demonstrated reduced mineralization of the callus, accompanied by elevated Sox9 expression and increased cartilage content, suggesting delayed repair. We also found that the chemokine Cxcl9 was highly upregulated in aged intact Prrx1+ pSSPCs, which has the potential to directly regulate other pSSPCs, and was associated with increased recruitment of CD8+ T cells at the fracture site. Cell-to-cell communication analysis provided further appreciation of the complex interactions among the many mesenchymal and hematopoietic cell types regulating fracture healing and highlighted the impact of aging on these interactions. Together, these results provide insight into age-induced alterations in early fracture healing, which could facilitate the development of improved therapeutic approaches for fracture repair in the elderly.
Collapse
Affiliation(s)
- Justin S King
- Department of Orthopedic Surgery, United States of America; UConn Musculoskeletal Institute, United States of America
| | - Matthew Wan
- Department of Orthopedic Surgery, United States of America; UConn Musculoskeletal Institute, United States of America
| | - Adam Kim
- Department of Medicine, United States of America
| | - Shagun Prabhu
- Department of Orthopedic Surgery, United States of America; UConn Musculoskeletal Institute, United States of America
| | - Sanja Novak
- UConn Musculoskeletal Institute, United States of America; Center for Regenerative Medicine and Skeletal Development, United States of America
| | - Ivo Kalajzic
- UConn Musculoskeletal Institute, United States of America; Center for Regenerative Medicine and Skeletal Development, United States of America
| | - Anne M Delany
- Department of Medicine, United States of America; Center for Molecular Oncology, UConn Health, Farmington, CT 06032, United States of America
| | - Archana Sanjay
- Department of Orthopedic Surgery, United States of America; UConn Musculoskeletal Institute, United States of America.
| |
Collapse
|
2
|
Ko FC, Fullam S, Lee H, Chan K, Ishihara S, Adamczyk NS, Obeidat AM, Soorya S, Miller RJ, Malfait AM, Miller RE. Clearing-enabled light sheet microscopy as a novel method for three-dimensional mapping of the sensory innervation of the mouse knee. J Orthop Res 2025; 43:632-639. [PMID: 39547819 PMCID: PMC11806991 DOI: 10.1002/jor.26016] [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: 09/09/2024] [Revised: 10/11/2024] [Accepted: 10/22/2024] [Indexed: 11/17/2024]
Abstract
A major barrier that hampers our understanding of the precise anatomic distribution of pain sensing nerves in and around the joint is the limited view obtained from traditional two dimensional (D) histological approaches. Therefore, our objective was to develop a workflow that allows examination of the innervation of the intact mouse knee joint in 3D by employing clearing-enabled light sheet microscopy. We first surveyed existing clearing protocols (SUMIC, PEGASOS, and DISCO) to determine their ability to clear the whole mouse knee joint, and discovered that a DISCO protocol provided the optimal transparency for light sheet microscopy imaging. We then modified the DISCO protocol to enhance binding and penetration of antibodies used for labeling nerves. Using the pan-neuronal PGP9.5 antibody, our protocol allowed 3D visualization of innervation in and around the mouse knee joint. We then implemented the workflow in mice intra-articularly injected with nerve growth factor (NGF) to determine whether changes in the nerve density can be observed. Both 3D and 2D analytical approaches of the light sheet microscopy images demonstrated quantifiable changes in midjoint nerve density following 4 weeks of NGF injection in the medial but not in the lateral joint compartment. We provide, for the first time, a comprehensive workflow that allows detailed and quantifiable examination of mouse knee joint innervation in 3D.
Collapse
Affiliation(s)
- Frank C. Ko
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Spencer Fullam
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL 60612, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Hoomin Lee
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL 60612, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Kelly Chan
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL 60612, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Shingo Ishihara
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL 60612, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Natalie S. Adamczyk
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL 60612, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Alia M. Obeidat
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL 60612, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Sarah Soorya
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL 60612, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Richard J. Miller
- Department of Pharmacology, Northwestern University, Chicago, IL 60611, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Anne-Marie Malfait
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL 60612, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Rachel E. Miller
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL 60612, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| |
Collapse
|
3
|
Horenberg AL, Ren Y, Zeng EZ, Rindone AN, Pathak AP, Grayson WL. 3D imaging reveals changes in the neurovascular architecture of the murine calvarium with aging. Bone Res 2025; 13:24. [PMID: 39984434 PMCID: PMC11845787 DOI: 10.1038/s41413-025-00401-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 12/18/2024] [Accepted: 12/31/2024] [Indexed: 02/23/2025] Open
Abstract
Calvarial nerves, along with vasculature, influence skull formation during development and following injury, but it remains unclear how calvarial nerves are spatially distributed during postnatal growth and aging. Studying the spatial distribution of nerves in the skull remains a challenge due to a lack of methods to quantify 3D structures in intact bone. To visualize calvarial 3D neurovascular architecture, we imaged nerves and endothelial cells with lightsheet microscopy. We employed machine-learning-based segmentation to facilitate high-resolution characterization from post-natal day 0 (P0) to 80 weeks. We found that TUBB3+ nerve density decreased with aging with the frontal bone demonstrating earlier onset age-related nerve loss than the parietal bone. In addition, nerves in the periosteum and dura mater exhibited similar yet distinct temporal patterns of nerve growth and loss. While no difference was observed in TUBB3+ nerves during skeletal maturation (P0 → 12 weeks), we did observe an increase in the volume of unmyelinated nerves in the dura mater. Regarding calvarial vasculature, larger CD31hiEmcn- vessel fraction increased with aging, while CD31hiEmcnhi vessel fraction was reduced. Throughout all ages, calvarial nerves maintained a preferential spatial association with CD31hiEmcnhi vessels, however, this association decreased with aging. Additionally, we used a model of Apert syndrome to explore the impact of suture-related disease on neurovascular architecture. Collectively, this 3D, spatiotemporal characterization of calvarial nerves throughout the lifespan and provides new insights into age-induced neurovascular architecture.
Collapse
Affiliation(s)
- Allison L Horenberg
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yunke Ren
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eric Z Zeng
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexandra N Rindone
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Arvind P Pathak
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Sciences, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Electrical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Warren L Grayson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
4
|
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
|
5
|
Li B, Ding Z, Ouchi T, Liao Y, Li B, Gong J, Xie Y, Zhao Z, Li L. Deciphering the spatial distribution of Gli1-lineage cells in dental, oral, and craniofacial regions. J Bone Miner Res 2024; 39:1809-1820. [PMID: 39303104 DOI: 10.1093/jbmr/zjae152] [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: 04/17/2024] [Revised: 09/11/2024] [Accepted: 09/18/2024] [Indexed: 09/22/2024]
Abstract
The craniofacial bone, crucial for protecting brain tissue and supporting facial structure, undergoes continuous remodeling through mesenchymal (MSCs) or skeletal stem cells in their niches. Gli1 is an ideal marker for labeling MSCs and osteoprogenitors in this region, and Gli1-lineage cells are identified as pivotal for bone growth, development, repair, and regeneration. Despite its significance, the distribution of Gli1-lineage cells across the dental, oral, and craniofacial (DOC) regions remains to be systematically explored. Utilizing tissue-clearing and light sheet fluorescence microscopy with a Gli1CreER; tdTomatoAi14 mouse model, we mapped the spatial distribution of Gli1-lineage cells throughout the skull, focusing on calvarial bones, sutures, bone marrow, teeth, periodontium, jaw bones, and the temporomandibular joint. We found Gli1-lineage cells widespread in these areas, underscoring their significance in DOC regions. Additionally, we observed their role in repairing calvarial bone defects, providing novel insights into craniofacial biology and stem cell niches and enhancing our understanding of stem cells and their progeny's behavior in vivo.
Collapse
Affiliation(s)
- Bo Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Zhangfan Ding
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Takehito Ouchi
- Department of Physiology, Tokyo Dental College, 2-9-18 Kanda-Misaki-cho, Chiyoda-ku, Tokyo 1010061, Japan
| | - Yueqi Liao
- Department of Biomedical Engineering, School of Big Health and Intelligent Engineering, Chengdu Medical College, Chengdu 610500, Sichuan, China
| | - Bingzhi Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Jiajing Gong
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yuhang Xie
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Longjiang Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
6
|
Li B. Guardians of the mind: Calvarial stem cells and brain border immunity. Stem Cell Reports 2024; 19:1520-1523. [PMID: 39486404 PMCID: PMC11589192 DOI: 10.1016/j.stemcr.2024.10.002] [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: 05/07/2024] [Revised: 10/03/2024] [Accepted: 10/03/2024] [Indexed: 11/04/2024] Open
Abstract
Calvarial bones safeguard the brain and are interconnected by immovable joints termed sutures, which function as growth centers for skull morphogenesis and stem cell niches. Recent years have witnessed paradigm shifts in this field, highlighting the essential roles of calvarial stem cells (CSCs), sutures, and surrounding structures in neuroimmune crosstalk and neurocognitive restoration.
Collapse
Affiliation(s)
- Bo Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, Ren Min Nan Road, Chengdu, Sichuan 610041, China.
| |
Collapse
|
7
|
King JS, Wan M, Kim A, Novak S, Prabhu S, Kalajzic I, Delany AM, Sanjay A. Effects of Aging on the Immune and Periosteal Response to Fracture in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.06.622348. [PMID: 39574733 PMCID: PMC11580938 DOI: 10.1101/2024.11.06.622348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
Aging predisposes individuals to reduced bone mass and fragility fractures, which are costly and linked to high mortality. Understanding how aging affects fracture healing is essential for developing therapies to enhance bone regeneration in older adults. During the inflammatory phase of fracture healing, immune cells are recruited to the injury site as periosteal skeletal stem/progenitor cells (pSSPCs) rapidly proliferate and differentiate into osteochondral lineages, allowing for fibrocartilaginous callus formation and complete bone healing. Irrespective of age, how periosteal mesenchymal and immune cells interact during early fracture healing is incompletely understood, limiting our ability to potentially modulate these processes. To address this, we directly analyzed, in parallel, at a single-cell level, isolated murine CD45(+) and CD45(-) periosteal cells dissected from intact and fractured bones, collected three days after injury. Through comprehensive analysis, corroborated by bulk RNA-sequencing, flow cytometry, and histology, we found aging decreases pSSPCs proliferative, marked by a reduced expression of genes required for callus formation and an increased senescence signature. We found that the chemokine Cxcl9 was highly upregulated in aged intact Prrx1+ pSSPCs, predicted to interact with other pSSPCs directly, and associated with increased recruitment of CD8+ T cells at the fracture site three days after injury. Cell-to-cell communication analysis provided insight into the complexity of interactions among the many cell types regulating fracture healing and the impact of aging on these processes. Together, these results provide insight into age-induced alterations in fracture healing, informing the development of improved therapeutic approaches for fragility fractures.
Collapse
|
8
|
Meslier QA, Duerr TJ, Guan W, Nguyen B, Monaghan JR, Shefelbine SJ. WISH-BONE: Whole-mount in situ histology, to label osteocyte mRNA and protein in 3D adult mouse bones. FASEB J 2024; 38:e70101. [PMID: 39387181 DOI: 10.1096/fj.202400635r] [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: 03/22/2024] [Revised: 07/19/2024] [Accepted: 08/20/2024] [Indexed: 10/12/2024]
Abstract
Bone is a three-dimensional (3D) highly dynamic tissue under constant remodeling. Commonly used tools to investigate bone biology require sample digestion for biomolecule extraction or provide only two-dimensional (2D) spatial information. There is a need for 3D tools to investigate spatially preserved biomarker expression in osteocytes. In this work, we present a new method, WISH-BONE, to label osteocyte messenger RNA (mRNA) and protein in whole-mount mouse bone. For mRNA labeling, we used hybridization chain reaction-fluorescence in situ hybridization (HCR-FISH) to label genes of interest in osteocytes. For protein labeling, samples were preserved using an epoxy-based solution that protects tissue structure and biomolecular components. Then an enzymatic matrix permeabilization step was performed to enable antibody penetration. Immunostaining was used to label various proteins involved in bone homeostasis. We also demonstrate the use of customized fluorescent nanobodies to target and label proteins in the cortical bone (CB). However, the relatively dim signal observed from nanobodies' staining limited detection. mRNA and protein labeling were performed in separate samples. In this study, we share protocols, highlight opportunities, and identify the challenges of this novel 3D labeling method. They are the first protocols for whole-mount osteocyte 3D labeling of mRNA and protein in mature mouse bones. WISH-BONE will allow the investigation of molecular signaling in bone cells in their 3D environment and could be applied to various bone-related fields of research.
Collapse
Affiliation(s)
- Quentin A Meslier
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
- LifeCanvas Technologies, Cambridge, Massachusetts, USA
| | - Timothy J Duerr
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
- Institute for Chemical Imaging of Living Systems, Northeastern University, Boston, Massachusetts, USA
| | - Webster Guan
- LifeCanvas Technologies, Cambridge, Massachusetts, USA
| | - Brian Nguyen
- LifeCanvas Technologies, Cambridge, Massachusetts, USA
| | - James R Monaghan
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
- Institute for Chemical Imaging of Living Systems, Northeastern University, Boston, Massachusetts, USA
| | - Sandra J Shefelbine
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
- Institute for Chemical Imaging of Living Systems, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Zanluqui NG, McGavern DB. Why do central nervous system barriers host a diverse immune landscape? Trends Immunol 2024; 45:738-749. [PMID: 39299888 PMCID: PMC11471389 DOI: 10.1016/j.it.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/22/2024]
Abstract
The meninges in vertebrates comprise three layers (dura, arachnoid, pia mater), representing an important barrier surrounding and protecting the central nervous system (CNS). The most exterior CNS barrier, the dura mater, is unique because it resembles a peripheral tissue. It hosts a rich immune landscape, lymphatic vessels, and fenestrated vasculature, allowing microbes and other threats from the blood to extravasate into the meninges, potentially reaching the underlying CNS. The highly specialized large venous drainage system in the dura is especially susceptible to infection. Here, we explore specializations in the CNS barrier system from an anatomical and immunological perspective and posit that the dura mater evolved an elaborate innate and adaptive immune system in specific locations within it to protect underlying CNS tissue against invading pathogens.
Collapse
Affiliation(s)
- Nagela G Zanluqui
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institute of Health (NIH), Bethesda, MD, USA
| | - Dorian B McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institute of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
10
|
Bixel MG, Sivaraj KK, Timmen M, Mohanakrishnan V, Aravamudhan A, Adams S, Koh BI, Jeong HW, Kruse K, Stange R, Adams RH. Angiogenesis is uncoupled from osteogenesis during calvarial bone regeneration. Nat Commun 2024; 15:4575. [PMID: 38834586 PMCID: PMC11150404 DOI: 10.1038/s41467-024-48579-5] [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: 05/24/2023] [Accepted: 05/06/2024] [Indexed: 06/06/2024] Open
Abstract
Bone regeneration requires a well-orchestrated cellular and molecular response including robust vascularization and recruitment of mesenchymal and osteogenic cells. In femoral fractures, angiogenesis and osteogenesis are closely coupled during the complex healing process. Here, we show with advanced longitudinal intravital multiphoton microscopy that early vascular sprouting is not directly coupled to osteoprogenitor invasion during calvarial bone regeneration. Early osteoprogenitors emerging from the periosteum give rise to bone-forming osteoblasts at the injured calvarial bone edge. Microvessels growing inside the lesions are not associated with osteoprogenitors. Subsequently, osteogenic cells collectively invade the vascularized and perfused lesion as a multicellular layer, thereby advancing regenerative ossification. Vascular sprouting and remodeling result in dynamic blood flow alterations to accommodate the growing bone. Single cell profiling of injured calvarial bones demonstrates mesenchymal stromal cell heterogeneity comparable to femoral fractures with increase in cell types promoting bone regeneration. Expression of angiogenesis and hypoxia-related genes are slightly elevated reflecting ossification of a vascularized lesion site. Endothelial Notch and VEGF signaling alter vascular growth in calvarial bone repair without affecting the ossification progress. Our findings may have clinical implications for bone regeneration and bioengineering approaches.
Collapse
Affiliation(s)
- M Gabriele Bixel
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine and University of Münster, Faculty of Medicine, D-48149, Münster, Germany.
| | - Kishor K Sivaraj
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine and University of Münster, Faculty of Medicine, D-48149, Münster, Germany
| | - Melanie Timmen
- Department of Regenerative Musculoskeletal Medicine, Institute of Musculoskeletal Medicine, University Hospital Münster, D-48149, Münster, Germany
| | - Vishal Mohanakrishnan
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine and University of Münster, Faculty of Medicine, D-48149, Münster, Germany
| | - Anusha Aravamudhan
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine and University of Münster, Faculty of Medicine, D-48149, Münster, Germany
| | - Susanne Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine and University of Münster, Faculty of Medicine, D-48149, Münster, Germany
| | - Bong-Ihn Koh
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine and University of Münster, Faculty of Medicine, D-48149, Münster, Germany
| | - Hyun-Woo Jeong
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine and University of Münster, Faculty of Medicine, D-48149, Münster, Germany
- Max Planck Institute for Molecular Biomedicine, Sequencing Core Facility, D-48149, Münster, Germany
| | - Kai Kruse
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine and University of Münster, Faculty of Medicine, D-48149, Münster, Germany
- Max Planck Institute for Molecular Biomedicine, Bioinformatics Service Unit, D-48149, Münster, Germany
| | - Richard Stange
- Department of Regenerative Musculoskeletal Medicine, Institute of Musculoskeletal Medicine, University Hospital Münster, D-48149, Münster, Germany
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine and University of Münster, Faculty of Medicine, D-48149, Münster, Germany.
| |
Collapse
|
11
|
Liu Z, Liu H, Liu S, Li B, Liu Y, Luo E. SIRT1 activation promotes bone repair by enhancing the coupling of type H vessel formation and osteogenesis. Cell Prolif 2024; 57:e13596. [PMID: 38211965 PMCID: PMC11150139 DOI: 10.1111/cpr.13596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/10/2023] [Accepted: 12/18/2023] [Indexed: 01/13/2024] Open
Abstract
Bone repair is intricately correlated with vascular regeneration, especially of type H vessels. Sirtuin 1 (SIRT1) expression is closely associated with endothelial function and vascular regeneration; however, the role of SIRT1 in enhancing the coupling of type H vessel formation with osteogenesis to promote bone repair needs to be investigated. A co-culture system combining human umbilical vein endothelial cells and osteoblasts was constructed, and a SIRT1 agonist was used to evaluate the effects of SIRT1 activity. The angiogenic and osteogenic capacities of the co-culture system were examined using short interfering RNA. Mouse models with bone defects in the femur or mandible were established to explore changes in type H vessel formation and bone repair following modulated SIRT1 activity. SIRT1 activation augmented the angiogenic and osteogenic capacities of the co-culture system by activating the PI3K/AKT/FOXO1 signalling pathway and did not significantly regulate osteoblast differentiation. Inhibition of the PI3K/AKT/FOXO1 pathway attenuated SIRT1-mediated effects. The SIRT1 activity in bone defects was positively correlated with the formation of type H vessels and bone repair in vivo, whereas SIRT1 inhibition substantially weakened vascular and bone formation. Thus, SIRT1 is crucial to the coupling of type H vessels with osteogenesis during bone repair.
Collapse
Affiliation(s)
- Zhikai Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Hanghang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Shibo Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Bolun Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yao Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - En Luo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
12
|
Ko FC, Fullam S, Lee H, Ishihara S, Adamczyk NS, Obeidat AM, Soorya S, Miller RJ, Malfait AM, Miller RE. Clearing-enabled light sheet microscopy as a novel method for three-dimensional mapping of the sensory innervation of the mouse knee. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.28.596316. [PMID: 38853939 PMCID: PMC11160612 DOI: 10.1101/2024.05.28.596316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
A major barrier that hampers our understanding of the precise anatomic distribution of pain sensing nerves in and around the joint is the limited view obtained from traditional two dimensional (D) histological approaches. Therefore, our objective was to develop a workflow that allows examination of the innervation of the intact mouse knee joint in 3D by employing clearing-enabled light sheet microscopy. We first surveyed existing clearing protocols (SUMIC, PEGASOS, and DISCO) to determine their ability to clear the whole mouse knee joint, and discovered that a DISCO protocol provided the most optimal transparency for light sheet microscopy imaging. We then modified the DISCO protocol to enhance binding and penetration of antibodies used for labeling nerves. Using the pan-neuronal PGP9.5 antibody, our protocol allowed 3D visualization of innervation in and around the mouse knee joint. We then implemented the workflow in mice intra-articularly injected with nerve growth factor (NGF) to determine whether changes in the nerve density can be observed. Both 3D and 2D analytical approaches of the light sheet microscopy images demonstrated quantifiable changes in midjoint nerve density following 4 weeks of NGF injection in the medial but not in the lateral joint compartment. We provide, for the first time, a comprehensive workflow that allows detailed and quantifiable examination of mouse knee joint innervation in 3D.
Collapse
Affiliation(s)
- Frank C Ko
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Spencer Fullam
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Hoomin Lee
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Shingo Ishihara
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Natalie S. Adamczyk
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Alia M. Obeidat
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Sarah Soorya
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Richard J. Miller
- Department of Pharmacology, Northwestern University, Chicago, IL 60611, USA
| | - Anne-Marie Malfait
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Rachel E. Miller
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
13
|
Perin P, Cossellu D, Vivado E, Batti L, Gantar I, Voigt FF, Pizzala R. Temporal bone marrow of the rat and its connections to the inner ear. Front Neurol 2024; 15:1386654. [PMID: 38817550 PMCID: PMC11137668 DOI: 10.3389/fneur.2024.1386654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/10/2024] [Indexed: 06/01/2024] Open
Abstract
Calvarial bone marrow has been found to be central in the brain immune response, being connected to the dura through channels which allow leukocyte trafficking. Temporal bone marrow is thought to play important roles in relation to the inner ear, but is still largely uncharacterized, given this bone complex anatomy. We characterized the geometry and connectivity of rat temporal bone marrow using lightsheet imaging of cleared samples and microCT. Bone marrow was identified in cleared tissue by cellular content (and in particular by the presence of megakaryocytes); since air-filled cavities are absent in rodents, marrow clusters could be recognized in microCT scans by their geometry. In cleared petrosal bone, autofluorescence allowed delineation of the otic capsule layers. Within the endochondral layer, bone marrow was observed in association to the cochlear base and vestibule, and to the cochlear apex. Cochlear apex endochondral marrow (CAEM) was a separated cluster from the remaining endochondral marrow, which was therefore defined as "vestibular endochondral marrow" (VEM). A much larger marrow island (petrosal non-endochondral marrow, PNEM) extended outside the otic capsule surrounding semicircular canal arms. PNEM was mainly connected to the dura, through bone channels similar to those of calvarial bone, and only a few channels were directed toward the canal periosteum. On the contrary, endochondral bone marrow was well connected to the labyrinth through vascular loops (directed to the spiral ligament for CAEM and to the bony labyrinth periosteum for VEM), and to dural sinuses. In addition, CAEM was also connected to the tensor tympani fossa of the middle ear and VEM to the endolymphatic sac. Endochondral marrow was made up of small lobules connected to each other and to other structures by channels lined by elongated macrophages, whereas PNEM displayed larger lobules connected by channels with a sparse macrophage population. Our data suggest that the rat inner ear is surrounded by bone marrow at the junctions with middle ear and brain, most likely with "customs" role, restricting pathogen spread; a second marrow network with different structural features is found within the endochondral bone layer of the otic capsule and may play different functional roles.
Collapse
Affiliation(s)
- Paola Perin
- Department of Brain and Behaviour Sciences, University of Pavia, Pavia, Italy
| | - Daniele Cossellu
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Elisa Vivado
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Laura Batti
- Wyss Center for Bio and Neuro Engineering, Geneva, Switzerland
| | - Ivana Gantar
- Wyss Center for Bio and Neuro Engineering, Geneva, Switzerland
| | - Fabian F. Voigt
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, United States
| | - Roberto Pizzala
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| |
Collapse
|
14
|
Ye J, Hua Z, Xiao J, Shao Y, Li S, Yin H, Wu M, Rong Y, Hong B, Guo Y, Ma Y, Wang J. p-Smad3 differentially regulates the cytological behavior of osteoclasts before and after osteoblasts maturation. Mol Biol Rep 2024; 51:525. [PMID: 38632128 DOI: 10.1007/s11033-024-09400-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 02/28/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND A series of previous investigations have revealed that p-Smad3 plays a facilitative role in the differentiation and maturation of osteoblasts, while also regulating the expression of certain intercellular communication factors. However, the effects of p-Smad3 in osteoblasts before and after maturation on the proliferation, migration, differentiation, apoptosis and other cellular behaviors of osteoclasts have not been reported. METHODS MC3T3-E1 cells were cultured in osteogenic induction medium for varying durations, After that, the corresponding conditioned medium was collected and the osteoclast lineage cells were treated. To elucidate the regulatory role of p-Smad3 within osteoblasts, we applied the activator TGF-β1 and inhibitor SIS3 to immature and mature osteoblasts and collected corresponding conditioned media for osteoclast intervention. RESULTS We observed an elevation of p-Smad3 and Smad3 during the early stage of osteoblast differentiation, followed by a decline in the later stage. we discovered that as osteoblasts mature, their conditioned media inhibit osteoclasts differentiation and the osteoclast-coupled osteogenic effect. However, it promotes apoptosis in osteoclasts and the angiogenesis coupled with osteoclasts. p-Smad3 in immature osteoblasts, through paracrine effects, promotes the migration, differentiation, and osteoclast-coupled osteogenic effects of osteoclast lineage cells. For mature osteoblasts, p-Smad3 facilitates osteoclast apoptosis and the angiogenesis coupled with osteoclasts. CONCLUSIONS As pre-osteoblasts undergo maturation, p-Smad3 mediated a paracrine effect that transitions osteoclast cellular behaviors from inducing differentiation and stimulating bone formation to promoting apoptosis and coupling angiogenesis.
Collapse
Affiliation(s)
- Jiapeng Ye
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, 214071, China
| | - Zhen Hua
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, 214071, China
| | - Jirimutu Xiao
- Laboratory of New Techniques of Restoration & Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
- Mongolian Medicine College, Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010159, China
| | - Yang Shao
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, 214071, China
| | - Shaoshuo Li
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, 214071, China
| | - Heng Yin
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, 214071, China
| | - Mao Wu
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, 214071, China
| | - Yi Rong
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, 214071, China
| | - Bowen Hong
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, 214071, China
| | - Yang Guo
- Laboratory of New Techniques of Restoration & Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Yong Ma
- Laboratory of New Techniques of Restoration & Reconstruction of Orthopedics and Traumatology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Jianwei Wang
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, Jiangsu, 214071, China.
| |
Collapse
|
15
|
Ren Y, Chu X, Senarathna J, Bhargava A, Grayson WL, Pathak AP. Multimodality imaging reveals angiogenic evolution in vivo during calvarial bone defect healing. Angiogenesis 2024; 27:105-119. [PMID: 38032405 PMCID: PMC10964991 DOI: 10.1007/s10456-023-09899-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023]
Abstract
The healing of calvarial bone defects is a pressing clinical problem that involves the dynamic interplay between angiogenesis and osteogenesis within the osteogenic niche. Although structural and functional vascular remodeling (i.e., angiogenic evolution) in the osteogenic niche is a crucial modulator of oxygenation, inflammatory and bone precursor cells, most clinical and pre-clinical investigations have been limited to characterizing structural changes in the vasculature and bone. Therefore, we developed a new multimodality imaging approach that for the first time enabled the longitudinal (i.e., over four weeks) and dynamic characterization of multiple in vivo functional parameters in the remodeled vasculature and its effects on de novo osteogenesis, in a preclinical calvarial defect model. We employed multi-wavelength intrinsic optical signal (IOS) imaging to assess microvascular remodeling, intravascular oxygenation (SO2), and osteogenesis; laser speckle contrast (LSC) imaging to assess concomitant changes in blood flow and vascular maturity; and micro-computed tomography (μCT) to validate volumetric changes in calvarial bone. We found that angiogenic evolution was tightly coupled with calvarial bone regeneration and corresponded to distinct phases of bone healing, such as injury, hematoma formation, revascularization, and remodeling. The first three phases occurred during the initial two weeks of bone healing and were characterized by significant in vivo changes in vascular morphology, blood flow, oxygenation, and maturity. Overall, angiogenic evolution preceded osteogenesis, which only plateaued toward the end of bone healing (i.e., four weeks). Collectively, these data indicate the crucial role of angiogenic evolution in osteogenesis. We believe that such multimodality imaging approaches have the potential to inform the design of more efficacious tissue-engineering calvarial defect treatments.
Collapse
Affiliation(s)
- Yunke Ren
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xinying Chu
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Janaka Senarathna
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 720 Rutland Ave, 217 Traylor Bldg, Baltimore, MD, 21205, USA
- Kavli Neuroscience Discovery Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Akanksha Bhargava
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 720 Rutland Ave, 217 Traylor Bldg, Baltimore, MD, 21205, USA
| | - Warren L Grayson
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Materials Science and Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, USA
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Arvind P Pathak
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 720 Rutland Ave, 217 Traylor Bldg, Baltimore, MD, 21205, USA.
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Electrical Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
16
|
Yeo M, Sarkar A, Singh YP, Derman ID, Datta P, Ozbolat IT. Synergistic coupling between 3D bioprinting and vascularization strategies. Biofabrication 2023; 16:012003. [PMID: 37944186 PMCID: PMC10658349 DOI: 10.1088/1758-5090/ad0b3f] [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: 01/19/2023] [Revised: 09/27/2023] [Accepted: 11/09/2023] [Indexed: 11/12/2023]
Abstract
Three-dimensional (3D) bioprinting offers promising solutions to the complex challenge of vascularization in biofabrication, thereby enhancing the prospects for clinical translation of engineered tissues and organs. While existing reviews have touched upon 3D bioprinting in vascularized tissue contexts, the current review offers a more holistic perspective, encompassing recent technical advancements and spanning the entire multistage bioprinting process, with a particular emphasis on vascularization. The synergy between 3D bioprinting and vascularization strategies is crucial, as 3D bioprinting can enable the creation of personalized, tissue-specific vascular network while the vascularization enhances tissue viability and function. The review starts by providing a comprehensive overview of the entire bioprinting process, spanning from pre-bioprinting stages to post-printing processing, including perfusion and maturation. Next, recent advancements in vascularization strategies that can be seamlessly integrated with bioprinting are discussed. Further, tissue-specific examples illustrating how these vascularization approaches are customized for diverse anatomical tissues towards enhancing clinical relevance are discussed. Finally, the underexplored intraoperative bioprinting (IOB) was highlighted, which enables the direct reconstruction of tissues within defect sites, stressing on the possible synergy shaped by combining IOB with vascularization strategies for improved regeneration.
Collapse
Affiliation(s)
- Miji Yeo
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, United States of America
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, United States of America
| | - Anwita Sarkar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal 700054, India
| | - Yogendra Pratap Singh
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, United States of America
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, United States of America
| | - Irem Deniz Derman
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, United States of America
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, United States of America
| | - Pallab Datta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal 700054, India
| | - Ibrahim T Ozbolat
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, United States of America
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, United States of America
- Department of Biomedical Engineering, Penn State University, University Park, PA 16802, United States of America
- Materials Research Institute, Penn State University, University Park, PA 16802, United States of America
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA 17033, United States of America
- Penn State Cancer Institute, Penn State University, Hershey, PA 17033, United States of America
- Biotechnology Research and Application Center, Cukurova University, Adana 01130, Turkey
| |
Collapse
|
17
|
Chen Y, Yin Y, Luo M, Wu J, Chen A, Deng L, Xie L, Han X. Occlusal Force Maintains Alveolar Bone Homeostasis via Type H Angiogenesis. J Dent Res 2023; 102:1356-1365. [PMID: 37786932 DOI: 10.1177/00220345231191745] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2023] Open
Abstract
Physiologically, teeth and periodontal tissues are exposed to occlusal forces throughout their lifetime. Following occlusal unloading, unbalanced bone remodeling manifests as a net alveolar bone (AB) loss. This phenomenon is termed alveolar bone disuse osteoporosis (ABDO), the underlying mechanism of which remains unclear. Type H vessels, a novel capillary subtype tightly coupled with osteogenesis, reportedly have a role in skeletal remodeling; however, their role in ABDO is not well studied. In the present study, we aimed to explore the pathogenesis of and therapies for ABDO. The study revealed that type H endothelium highly positive for CD31 and endomucin was identified in the periodontal ligament (PDL) but rarely in the AB of the mice. In hypofunctional PDL, the density of type H vasculature and coupled osterix+ (OSX+) osteoprogenitors declined significantly. In addition, the angiogenic factor Slit guidance ligand 3 (SLIT3) was downregulated in the disused PDL, and periodontal injection of the recombinant SLIT3 protein partially ameliorated type H vessel dysfunction and AB loss in ABDO mice. With regard to the molecular mechanism, a mechanosensory signaling circuit, PIEZO1/Ca2+/HIF-1α/SLIT3, was validated by applying cyclic compression to 3-dimensional-cultured PDL cells using the Flexcell FX-5000 compression system. In summary, PDL plays a pivotal role in mechanotransduction by translating physical forces into the intracellular signaling axis PIEZO1/Ca2+/HIF-1α/SLIT3, which promotes type H angiogenesis and OSX+ cell-related osteogenensis, thereby contributing to AB homeostasis. Our findings advance the understanding of PDL in AB disorders. Further therapies targeting SLIT3 may provide new insights into preventing bone loss in ABDO.
Collapse
Affiliation(s)
- Y Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Y Yin
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - M Luo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - J Wu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - A Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - L Deng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - L Xie
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - X Han
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
18
|
Sun H, Meng S, Xu Z, Cai H, Pei X, Wan Q, Chen J. Vascular and lymphatic heterogeneity and age-related variations of dental pulps. J Dent 2023; 138:104695. [PMID: 37714450 DOI: 10.1016/j.jdent.2023.104695] [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: 07/01/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/17/2023] Open
Abstract
OBJECTIVES Dental pulp tissue is highly vascularized. However, age-related vascular changes of the dental pulp in mice and humans remain poorly understood. We modified a novel tissue clearing method, mapped the vasculature, pericytes, and perivascular matrix in the dental pulp via high-resolution 3D imaging. METHODS We isolated young and aged pulps from mouse teeth, and mapped vasculature through a high-resolution thick frozen sections imaging method and a modified tissue clearing method. Human dental pulps were also mapped for vasculature studying. Furthermore, young and aged human dental pulps were collected and were compared with mouse pulps through RNA- sequencing. RESULTS Five vascular subtypes of blood vessels were found in the mouse dental pulp, which constituted the arterioles-capillaries-venules network. The density of capillaries and venules of molars declined obviously in aged mice. Among the age-dependent changes in the perivascular pulp matrix, the perivascular macrophages remarkably increased, lymphatic capillaries increased, while the nerves and extracellular matrix remained unchanged. Furthermore, the vascular patterns of human formed a complex vascular network. Both mouse and human dental pulps exhibited an inflammaging state. TNF pathway and Rap1 pathway might become promising targets for combating inflammaging and promoting angiogenesis. CONCLUSIONS Five subtypes of blood vessels were identified within the dental pulp of mice. Notably, the density of capillaries and venules in pulps of aged mice was reduced. Furthermore, partial similarities were observed in the vascular patterns between the dental pulps of humans and mice. RNA-sequencing analysis revealed that both mouse and human dental pulps exhibit indications of an inflammaging state. CLINICAL SIGNIFICANCE This study may contribute to unraveling potential therapeutic targets in the pulp regeneration and treatment of relevant diseases in the elderly.
Collapse
Affiliation(s)
- Haiyang Sun
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shuhuai Meng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhengyi Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - He Cai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xibo Pei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qianbing Wan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Junyu Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
19
|
Li B, Li J, Li B, Ouchi T, Li L, Li Y, Zhao Z. A single-cell transcriptomic atlas characterizes age-related changes of murine cranial stem cell niches. Aging Cell 2023; 22:e13980. [PMID: 37681346 PMCID: PMC10652347 DOI: 10.1111/acel.13980] [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: 06/16/2023] [Revised: 08/21/2023] [Accepted: 08/21/2023] [Indexed: 09/09/2023] Open
Abstract
The craniofacial bones provide structural support for the skull and accommodate the vulnerable brain tissue with a protective cavity. The bone tissue undergoes constant turnover, which relies on skeletal stem cells (SSCs) and/or mesenchymal stem cells (MSCs) and their niches. SSCs/MSCs and their perivascular niche within the bone marrow are well characterized in long bones. As for cranial bones, besides bone marrow, the suture mesenchyme has been identified as a unique niche for SSCs/MSCs of craniofacial bones. However, a comprehensive study of the two different cranial stem cell niches at single-cell resolution is still lacking. In addition, during the progression of aging, age-associated changes in cranial stem cell niches and resident cells remain uncovered. In this study, we investigated age-related changes in cranial stem cell niches via single-cell RNA sequencing (scRNA-seq). The transcriptomic profiles and cellular compositions have been delineated, indicating alterations of the cranial bone marrow microenvironment influenced by inflammaging. Moreover, we identified a senescent mesenchymal cell subcluster and several age-related immune cell subclusters by reclustering and pseudotime trajectory analysis, which might be closely linked to inflammaging. Finally, differentially expressed genes (DEGs) and cell-cell communications were analyzed during aging, revealing potential regulatory factors. Overall, this work highlights the age-related changes in cranial stem cell niches, which deepens the current understanding of cranial bone and suture biology and may provide therapeutic targets for antiaging and regenerative medicine.
Collapse
Affiliation(s)
- Bo Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of StomatologySichuan UniversitySichuanChengduChina
| | - Jingya Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of StomatologySichuan UniversitySichuanChengduChina
| | - Bingzhi Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of StomatologySichuan UniversitySichuanChengduChina
| | | | - Longjiang Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of StomatologySichuan UniversitySichuanChengduChina
| | - Yu Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of StomatologySichuan UniversitySichuanChengduChina
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of StomatologySichuan UniversitySichuanChengduChina
| |
Collapse
|
20
|
Li B, Li J, Fan Y, Zhao Z, Li L, Okano H, Ouchi T. Dissecting calvarial bones and sutures at single-cell resolution. Biol Rev Camb Philos Soc 2023; 98:1749-1767. [PMID: 37171117 DOI: 10.1111/brv.12975] [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: 02/10/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 05/13/2023]
Abstract
Cranial bones constitute a protective shield for the vulnerable brain tissue, bound together as a rigid entity by unique immovable joints known as sutures. Cranial sutures serve as major growth centres for calvarial morphogenesis and have been identified as a niche for mesenchymal stem cells (MSCs) and/or skeletal stem cells (SSCs) in the craniofacial skeleton. Despite the established dogma of cranial bone and suture biology, technological advancements now allow us to investigate these tissues and structures at unprecedented resolution and embrace multiple novel biological insights. For instance, a decrease or imbalance of representation of SSCs within sutures might underlie craniosynostosis; dural sinuses enable neuroimmune crosstalk and are newly defined as immune hubs; skull bone marrow acts as a myeloid cell reservoir for the meninges and central nervous system (CNS) parenchyma in mediating immune surveillance, etc. In this review, we revisit a growing body of recent studies that explored cranial bone and suture biology using cutting-edge techniques and have expanded our current understanding of this research field, especially from the perspective of development, homeostasis, injury repair, resident MSCs/SSCs, immunosurveillance at the brain's border, and beyond.
Collapse
Affiliation(s)
- Bo Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jingya Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yi Fan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Longjiang Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 1608582, Japan
- Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako-shi, Saitama, 3510198, Japan
| | - Takehito Ouchi
- Department of Physiology, Tokyo Dental College, 2-9-18 Kanda-Misaki-cho, Chiyoda-ku, Tokyo, 1010061, Japan
| |
Collapse
|
21
|
Buenaventura RG, Harvey AC, Burns MP, Main BS. Traumatic brain injury induces an adaptive immune response in the meningeal transcriptome that is amplified by aging. Front Neurosci 2023; 17:1210175. [PMID: 37588516 PMCID: PMC10425597 DOI: 10.3389/fnins.2023.1210175] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/07/2023] [Indexed: 08/18/2023] Open
Abstract
Traumatic Brain Injury (TBI) is a major cause of disability and mortality, particularly among the elderly, yet our mechanistic understanding of how age renders the post-traumatic brain vulnerable to poor clinical outcomes and susceptible to neurological disease remains poorly understood. It is well established that dysregulated and sustained immune responses contribute to negative outcomes after TBI, however our understanding of the interactions between central and peripheral immune reservoirs is still unclear. The meninges serve as the interface between the brain and the immune system, facilitating important bi-directional roles in healthy and disease settings. It has been previously shown that disruption of this system exacerbates inflammation in age related neurodegenerative disorders such as Alzheimer's disease, however we have an incomplete understanding of how the meningeal compartment influences immune responses after TBI. Here, we examine the meningeal tissue and its response to brain injury in young (3-months) and aged (18-months) mice. Utilizing a bioinformatic approach, high-throughput RNA sequencing demonstrates alterations in the meningeal transcriptome at sub-acute (7-days) and chronic (1 month) timepoints after injury. We find that age alone chronically exacerbates immunoglobulin production and B cell responses. After TBI, adaptive immune response genes are up-regulated in a temporal manner, with genes involved in T cell responses elevated sub-acutely, followed by increases in B cell related genes at chronic time points after injury. Pro-inflammatory cytokines are also implicated as contributing to the immune response in the meninges, with ingenuity pathway analysis identifying interferons as master regulators in aged mice compared to young mice following TBI. Collectively these data demonstrate the temporal series of meningeal specific signatures, providing insights into how age leads to worse neuroinflammatory outcomes in TBI.
Collapse
Affiliation(s)
| | | | | | - Bevan S. Main
- Laboratory for Brain Injury and Dementia, Department of Neuroscience, Georgetown University, Washington, DC, United States
| |
Collapse
|
22
|
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
|
23
|
Goertz JE, Garcia-Bonilla L, Iadecola C, Anrather J. Immune compartments at the brain's borders in health and neurovascular diseases. Semin Immunopathol 2023; 45:437-449. [PMID: 37138042 PMCID: PMC10279585 DOI: 10.1007/s00281-023-00992-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/14/2023] [Indexed: 05/05/2023]
Abstract
Recent evidence implicates cranial border immune compartments in the meninges, choroid plexus, circumventricular organs, and skull bone marrow in several neuroinflammatory and neoplastic diseases. Their pathogenic importance has also been described for cardiovascular diseases such as hypertension and stroke. In this review, we will examine the cellular composition of these cranial border immune niches, the potential pathways through which they might interact, and the evidence linking them to cardiovascular disease.
Collapse
Affiliation(s)
- Jennifer E Goertz
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61St Street; RR-405, New York, NY, 10065, USA
| | - Lidia Garcia-Bonilla
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61St Street; RR-405, New York, NY, 10065, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61St Street; RR-405, New York, NY, 10065, USA
| | - Josef Anrather
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61St Street; RR-405, New York, NY, 10065, USA.
| |
Collapse
|
24
|
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
|
25
|
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
|
26
|
Aldawood ZA, Mancinelli L, Geng X, Yeh SCA, Di Carlo R, C. Leite T, Gustafson J, Wilk K, Yozgatian J, Garakani S, Bassir SH, Cunningham ML, Lin CP, Intini G. Expansion of the sagittal suture induces proliferation of skeletal stem cells and sustains endogenous calvarial bone regeneration. Proc Natl Acad Sci U S A 2023; 120:e2120826120. [PMID: 37040407 PMCID: PMC10120053 DOI: 10.1073/pnas.2120826120] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/30/2023] [Indexed: 04/12/2023] Open
Abstract
In newborn humans, and up to approximately 2 y of age, calvarial bone defects can naturally regenerate. This remarkable regeneration potential is also found in newborn mice and is absent in adult mice. Since previous studies showed that the mouse calvarial sutures are reservoirs of calvarial skeletal stem cells (cSSCs), which are the cells responsible for calvarial bone regeneration, here we hypothesized that the regenerative potential of the newborn mouse calvaria is due to a significant amount of cSSCs present in the newborn expanding sutures. Thus, we tested whether such regenerative potential can be reverse engineered in adult mice by artificially inducing an increase of the cSSCs resident within the adult calvarial sutures. First, we analyzed the cellular composition of the calvarial sutures in newborn and in older mice, up to 14-mo-old mice, showing that the sutures of the younger mice are enriched in cSSCs. Then, we demonstrated that a controlled mechanical expansion of the functionally closed sagittal sutures of adult mice induces a significant increase of the cSSCs. Finally, we showed that if a calvarial critical size bone defect is created simultaneously to the mechanical expansion of the sagittal suture, it fully regenerates without the need for additional therapeutic aids. Using a genetic blockade system, we further demonstrate that this endogenous regeneration is mediated by the canonical Wnt signaling. This study shows that controlled mechanical forces can harness the cSSCs and induce calvarial bone regeneration. Similar harnessing strategies may be used to develop novel and more effective bone regeneration autotherapies.
Collapse
Affiliation(s)
- Zahra A. Aldawood
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA02115
- Department of Biomedical Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam34212, Saudi Arabia
| | - Luigi Mancinelli
- Department of Periodontics and Preventive Dentistry, University of PittsburghSchool of Dental Medicine, Pittsburgh, PA15261
- Center for Craniofacial Regeneration, University of PittsburghSchool of Dental Medicine, Pittsburgh, PA15261
| | - Xuehui Geng
- Department of Periodontics and Preventive Dentistry, University of PittsburghSchool of Dental Medicine, Pittsburgh, PA15261
- Center for Craniofacial Regeneration, University of PittsburghSchool of Dental Medicine, Pittsburgh, PA15261
| | - Shu-Chi A. Yeh
- Advanced Microscopy Program, Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA02114
| | - Roberta Di Carlo
- Department of Periodontics and Preventive Dentistry, University of PittsburghSchool of Dental Medicine, Pittsburgh, PA15261
- Center for Craniofacial Regeneration, University of PittsburghSchool of Dental Medicine, Pittsburgh, PA15261
| | - Taiana C. Leite
- Department of Periodontics and Preventive Dentistry, University of PittsburghSchool of Dental Medicine, Pittsburgh, PA15261
- Center for Craniofacial Regeneration, University of PittsburghSchool of Dental Medicine, Pittsburgh, PA15261
| | - Jonas Gustafson
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA98101
| | - Katarzyna Wilk
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA02115
| | - Joseph Yozgatian
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA02115
| | - Sasan Garakani
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA02115
| | - Seyed Hossein Bassir
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA02115
| | - Michael L. Cunningham
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA98101
- Division of Craniofacial Medicine, Department of Pediatrics, University of Washington, Seattle, WA98195
| | - Charles P. Lin
- Advanced Microscopy Program, Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA02114
| | - Giuseppe Intini
- Department of Periodontics and Preventive Dentistry, University of PittsburghSchool of Dental Medicine, Pittsburgh, PA15261
- Center for Craniofacial Regeneration, University of PittsburghSchool of Dental Medicine, Pittsburgh, PA15261
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA15261
- University of Pittsburgh UPMC Hillman Cancer Center, Pittsburgh, PA15232
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA15219
| |
Collapse
|
27
|
Li Y, Zhu J, Zhang X, Li Y, Zhang S, Yang L, Li R, Wan Q, Pei X, Chen J, Wang J. Drug-Delivery Nanoplatform with Synergistic Regulation of Angiogenesis-Osteogenesis Coupling for Promoting Vascularized Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2023; 15:17543-17561. [PMID: 37010447 DOI: 10.1021/acsami.2c23107] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
It has been confirmed that substantial vascularization is an effective strategy to heal large-scale bone defects in the field of bone tissue engineering. The local application of deferoxamine (DFO) is among the most common and effective methods for promoting the formation of blood vessels, although its short half-life in plasma, rapid clearance, and poor biocompatibility limit its therapeutic suitability. Herein, zeolitic imidazolate framework-8 (ZIF-8) was selected as a vehicle to extend the half-life of DFO. In the present study, a nano DFO-loaded ZIF-8 (DFO@ZIF-8) drug delivery system was established to promote angiogenesis-osteogenesis coupling. The nanoparticles were characterized, and their drug loading efficiency was examined to confirm the successful synthesis of nano DFO@ZIF-8. Additionally, due to the sustained release of DFO and Zn2+, DFO@ZIF-8 NPs were able to promote angiogenesis in human umbilical vein endothelial cells (HUVECs) culture and osteogenesis in bone marrow stem cells (BMSCs) in vitro. Furthermore, the DFO@ZIF-8 NPs promoted vascularization by enhancing the expression of type H vessels and a vascular network. The DFO@ZIF-8 NPs promoted bone regeneration in vivo by increasing the expression of OCN and BMP-2. RNA sequencing analysis revealed that the PI3K-AKT-MMP-2/9 and HIF-1α pathways were upregulated by DFO@ZIF-8 NPs in HUVECs, ultimately leading to the formation of new blood vessels. In addition, the mechanism by which DFO@ZIF-8 NPs promoted bone regeneration was potentially related to the synergistic effect of angiogenesis-osteogenesis coupling and Zn2+-mediation of the MAPK pathway. Taken together, DFO@ZIF-8 NPs, which were demonstrated to have low cytotoxicity and excellent coupling of angiogenesis and osteogenesis, represent a promising strategy for the reconstruction of critical-sized bone defects.
Collapse
Affiliation(s)
- Yahong Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Junjin Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xin Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yuanyuan Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Shu Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Linxin Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ruyi Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Qianbing Wan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xibo Pei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Junyu Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jian Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu 610041, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
28
|
Lu W, Zeng M, Liu W, Ma T, Fan X, Li H, Wang Y, Wang H, Hu Y, Xie J. Human urine-derived stem cell exosomes delivered via injectable GelMA templated hydrogel accelerate bone regeneration. Mater Today Bio 2023; 19:100569. [PMID: 36846309 PMCID: PMC9945756 DOI: 10.1016/j.mtbio.2023.100569] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 01/15/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
The key to critical bone regeneration in tissue engineering relies on an ideal bio-scaffold coated with a controlled release of growth factors. Gelatin methacrylate (GelMA) and Hyaluronic acid methacrylate (HAMA) have been a novel topic of interest in bone regeneration while introducing appropriate nano-hydroxyapatite (nHAP) to improve its mechanical properties. And the exosomes derived from human urine-derived stem cells (human USCEXOs) have also been reported to promote osteogenesis in tissue engineering. The present study aimed to design a new GelMA-HAMA/nHAP composite hydrogel as a drug delivery system. The USCEXOs were encapsulated and slow-released in the hydrogel for better osteogenesis. The characterization of the GelMA-based hydrogel showed excellent controlled release performance and appropriate mechanical properties. The in vitro studies showed that the USCEXOs/GelMA-HAMA/nHAP composite hydrogel could promote the osteogenesis of bone marrow mesenchymal stem cells (BMSCs) and the angiogenesis of endothelial progenitor cells (EPCs), respectively. Meanwhile, the in vivo results confirmed that this composite hydrogel could significantly promote the defect repair of cranial bone in the rat model. In addition, we also found that USCEXOs/GelMA-HAMA/nHAP composite hydrogel can promote the formation of H-type vessels in the bone regeneration area, enhancing the therapeutic effect. In conclusion, our findings suggested that this controllable and biocompatible USCEXOs/GelMA-HAMA/nHAP composite hydrogel may effectively promote bone regeneration by coupling osteogenesis and angiogenesis.
Collapse
Affiliation(s)
- Wei Lu
- Department of Orthopedic Surgery, National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Changsha, China
| | - Min Zeng
- Department of Orthopedic Surgery, National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Changsha, China
| | - Wenbin Liu
- Department of Orthopedic Surgery, National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Changsha, China
| | - Tianliang Ma
- Department of Orthopedic Surgery, National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Changsha, China
| | - Xiaolei Fan
- Department of Orthopedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Hui Li
- Department of Orthopedics, The First Affiliated Hospital, Medical College of Zhejiang University, Hangzhou, China
| | - Yinan Wang
- Department of Orthopedic Surgery, National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Changsha, China
| | - Haoyi Wang
- Department of Orthopedic Surgery, National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Changsha, China
| | - Yihe Hu
- Department of Orthopedic Surgery, National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Orthopedics, The First Affiliated Hospital, Medical College of Zhejiang University, Hangzhou, China
| | - Jie Xie
- Department of Orthopedic Surgery, National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Orthopedics, The First Affiliated Hospital, Medical College of Zhejiang University, Hangzhou, China
| |
Collapse
|
29
|
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
|
30
|
Timberlake AT, Kiziltug E, Jin SC, Nelson-Williams C, Loring E, Allocco A, Marlier A, Banka S, Stuart H, Passos-Buenos MR, Rosa R, Rogatto SR, Tonne E, Stiegler AL, Boggon TJ, Alperovich M, Steinbacher D, Staffenberg DA, Flores RL, Persing JA, Kahle KT, Lifton RP. De novo mutations in the BMP signaling pathway in lambdoid craniosynostosis. Hum Genet 2023; 142:21-32. [PMID: 35997807 DOI: 10.1007/s00439-022-02477-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/08/2022] [Indexed: 01/18/2023]
Abstract
Lambdoid craniosynostosis (CS) is a congenital anomaly resulting from premature fusion of the cranial suture between the parietal and occipital bones. Predominantly sporadic, it is the rarest form of CS and its genetic etiology is largely unexplored. Exome sequencing of 25 kindreds, including 18 parent-offspring trios with sporadic lambdoid CS, revealed a marked excess of damaging (predominantly missense) de novo mutations that account for ~ 40% of sporadic cases. These mutations clustered in the BMP signaling cascade (P = 1.6 × 10-7), including mutations in genes encoding BMP receptors (ACVRL1 and ACVR2A), transcription factors (SOX11, FOXO1) and a transcriptional co-repressor (IFRD1), none of which have been implicated in other forms of CS. These missense mutations are at residues critical for substrate or target sequence recognition and many are inferred to cause genetic gain-of-function. Additionally, mutations in transcription factor NFIX were implicated in syndromic craniosynostosis affecting diverse sutures. Single cell RNA sequencing analysis of the mouse lambdoid suture identified enrichment of mutations in osteoblast precursors (P = 1.6 × 10-6), implicating perturbations in the balance between proliferation and differentiation of osteoprogenitor cells in lambdoid CS. The results contribute to the growing knowledge of the genetics of CS, have implications for genetic counseling, and further elucidate the molecular etiology of premature suture fusion.
Collapse
Affiliation(s)
- Andrew T Timberlake
- Hansjörg Wyss Department of Plastic Surgery, New York University Langone Medical Center, New York, NY, USA.
| | - Emre Kiziltug
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Sheng Chih Jin
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.,Department of Genetics, Washington University School of Medicine, St Louis, MO, USA
| | | | - Erin Loring
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | | | - August Allocco
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, USA
| | - Arnaud Marlier
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, USA
| | - Siddharth Banka
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9WL, UK.,Manchester Centre for Genomic Medicine, Health Innovation Manchester, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, M13 9WL, UK
| | - Helen Stuart
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9WL, UK.,Manchester Centre for Genomic Medicine, Health Innovation Manchester, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, M13 9WL, UK
| | | | - Rafael Rosa
- Clinical Genetics, UFCSPA and Irmandade da Santa Casa de Misericórdia de Porto Alegre (ISCMPA), Porto Alegre, RS, Brazil
| | - Silvia R Rogatto
- Neogene Laboratory, Research Center (CIPE), AC Camargo Cancer Center, São Paulo, SP, Brazil
| | - Elin Tonne
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway.,University of Oslo, Oslo, Norway
| | - Amy L Stiegler
- Department of Pharmacology, Yale University, New Haven, CT, USA
| | - Titus J Boggon
- Department of Pharmacology, Yale University, New Haven, CT, USA
| | - Michael Alperovich
- Section of Plastic and Reconstructive Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Derek Steinbacher
- Section of Plastic and Reconstructive Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - David A Staffenberg
- Hansjörg Wyss Department of Plastic Surgery, New York University Langone Medical Center, New York, NY, USA
| | - Roberto L Flores
- Hansjörg Wyss Department of Plastic Surgery, New York University Langone Medical Center, New York, NY, USA
| | - John A Persing
- Section of Plastic and Reconstructive Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Kristopher T Kahle
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,Broad Institute of Harvard and Massachusetts Institute of Technology, Boston, MA, USA.,Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
| | - Richard P Lifton
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA. .,Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
31
|
Shirai Y, Okano J, Nakagawa T, Katagi M, Nakae Y, Arakawa A, Koshinuma S, Yamamoto G, Kojima H. Bone marrow-derived vasculogenesis leads to scarless regeneration in deep wounds with periosteal defects. Sci Rep 2022; 12:20589. [PMID: 36446886 PMCID: PMC9708684 DOI: 10.1038/s41598-022-24957-1] [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: 10/11/2022] [Accepted: 11/22/2022] [Indexed: 12/02/2022] Open
Abstract
Deep skin wounds with periosteal defects, frequently caused by traffic accidents or radical dissection, are refractory. Transplant surgery is frequently performed, but patients are subjected to stress for long operation periods, the sacrifice of donor regions, or several complications, such as flap necrosis or intractable ulcers. Even if the defects are covered, a scar composed of fibrous tissue remains in the body, which can cause itching, dysesthesia, or repeated ulcers because of the lack of distribution of peripheral nerves or hair follicles. Thus, treatments with the aim of regenerating lost tissue for deep wounds with periosteal defects are needed. Here, we show that the use of gelatin sponges (GS), which have been used as haemostatic materials in clinical practice, allowed the regeneration of heterogeneous tissues, including periosteum, skin, and skin appendages, when used as scaffolds in deep wounds with periosteal defects in rats. Bone marrow transplantation in rats revealed the mechanism by which the microenvironment provided by GS enabled bone marrow-derived cells (BMDCs) to form a vascular niche, followed by regeneration of the periosteum, skin, or skin appendages such as hair follicles by local cells. Our findings demonstrated that vascular niche formation provided by BMDCs is crucial for heterogeneous tissue regeneration.
Collapse
Affiliation(s)
- Yuuki Shirai
- grid.410827.80000 0000 9747 6806Department of Oral and Maxillofacial Surgery, Shiga University of Medical Science, Shiga, Japan
| | - Junko Okano
- grid.410827.80000 0000 9747 6806Department of Plastic and Reconstructive Surgery, Shiga University of Medical Science, Shiga, Japan
| | - Takahiko Nakagawa
- grid.410827.80000 0000 9747 6806Department of Regenerative Medicine Development, Shiga University of Medical Science, Shiga, Japan ,grid.410827.80000 0000 9747 6806Department of Biocommunication Development, Shiga University of Medical Science, Shiga, Japan
| | - Miwako Katagi
- grid.410827.80000 0000 9747 6806Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Shiga, Japan
| | - Yuki Nakae
- grid.410827.80000 0000 9747 6806Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Shiga, Japan
| | - Atsuhiro Arakawa
- grid.410827.80000 0000 9747 6806Department of Plastic and Reconstructive Surgery, Shiga University of Medical Science, Shiga, Japan
| | - Shinya Koshinuma
- grid.410827.80000 0000 9747 6806Department of Oral and Maxillofacial Surgery, Shiga University of Medical Science, Shiga, Japan
| | - Gaku Yamamoto
- grid.410827.80000 0000 9747 6806Department of Oral and Maxillofacial Surgery, Shiga University of Medical Science, Shiga, Japan
| | - Hideto Kojima
- grid.410827.80000 0000 9747 6806Department of Regenerative Medicine Development, Shiga University of Medical Science, Shiga, Japan ,grid.410827.80000 0000 9747 6806Department of Biocommunication Development, Shiga University of Medical Science, Shiga, Japan
| |
Collapse
|
32
|
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
|
33
|
Ren Y, Senarathna J, Grayson WL, Pathak AP. State-of-the-art techniques for imaging the vascular microenvironment in craniofacial bone tissue engineering applications. Am J Physiol Cell Physiol 2022; 323:C1524-C1538. [PMID: 36189973 PMCID: PMC9829486 DOI: 10.1152/ajpcell.00195.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 09/07/2022] [Accepted: 09/27/2022] [Indexed: 01/21/2023]
Abstract
Vascularization is a crucial step during musculoskeletal tissue regeneration via bioengineered constructs or grafts. Functional vasculature provides oxygen and nutrients to the graft microenvironment, facilitates wound healing, enhances graft integration with host tissue, and ensures the long-term survival of regenerating tissue. Therefore, imaging de novo vascularization (i.e., angiogenesis), changes in microvascular morphology, and the establishment and maintenance of perfusion within the graft site (i.e., vascular microenvironment or VME) can provide essential insights into engraftment, wound healing, as well as inform the design of tissue engineering (TE) constructs. In this review, we focus on state-of-the-art imaging approaches for monitoring the VME in craniofacial TE applications, as well as future advances in this field. We describe how cutting-edge in vivo and ex vivo imaging methods can yield invaluable information regarding VME parameters that can help characterize the effectiveness of different TE constructs and iteratively inform their design for enhanced craniofacial bone regeneration. Finally, we explicate how the integration of novel TE constructs, preclinical model systems, imaging techniques, and systems biology approaches could usher in an era of "image-based tissue engineering."
Collapse
Affiliation(s)
- Yunke Ren
- Department of Biomedical Engineering, the Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Janaka Senarathna
- Russell H. Morgan Department of Radiology and Radiological Sciences, the Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Warren L Grayson
- Department of Biomedical Engineering, the Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, Maryland
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland
| | - Arvind P Pathak
- Russell H. Morgan Department of Radiology and Radiological Sciences, the Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Biomedical Engineering, the Johns Hopkins University School of Medicine, Baltimore, Maryland
- Sidney Kimmel Comprehensive Cancer Center, the Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Electrical Engineering, Johns Hopkins University, Baltimore, Maryland
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
34
|
Zeng Y, Huang C, Duan D, Lou A, Guo Y, Xiao T, Wei J, Liu S, Wang Z, Yang Q, Zhou L, Wu Z, Wang L. Injectable temperature-sensitive hydrogel system incorporating deferoxamine-loaded microspheres promotes H-type blood vessel-related bone repair of a critical size femoral defect. Acta Biomater 2022; 153:108-123. [PMID: 36115651 DOI: 10.1016/j.actbio.2022.09.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 09/02/2022] [Accepted: 09/08/2022] [Indexed: 11/01/2022]
Abstract
Insufficient vascularization is a major challenge in the repair of critical-sized bone defects. Deferoxamine (DFO) has been reported to play a potential role in promoting the formation of H-type blood vessels, a specialized vascular subtype with coupled angiogenesis and osteogenesis. However, whether DFO promotes the expression of H-type vessels in critical femoral defects with complete periosteal damage remains unknown. Moreover, stable drug loading systems need to be designed owing to the short half-life and high-dose toxic effects of DFO. In this study, we developed an injectable DFO-gelatin microspheres (GMs) hydrogel complex as a stable drug loading system for the treatment of critical femoral defects in rats. Our results showed that sustained release of DFO in critical femoral defects stimulated the generation of functional H-type vessels. The DFO-GMs hydrogel complex effectively promoted proliferation, formation, and migration of human umbilical vein endothelial cells in vitro. In vivo, the application of the DFO-GMs hydrogel complex expanded the distribution range and prolonged the expression time of H-type vessels in the defect area and was positively correlated with the number of osterix+ cells and new bone tissue. Topical application of the HIF-1α inhibitor PX-478 partially blocked the stimulation of H-type vessels by DFO, whereas the osteogenic potential of the latter was also weakened. Our results extended the local application of DFO and provided a theoretical basis for targeting H-type vessels to treat large femoral defects. STATEMENT OF SIGNIFICANCE: Abundant functional blood vessels are essential for bone repair. The H-type blood vessel is a functional subtype with angiogenesis and osteogenesis coupling potential. A drug loading system with long-term controlled release was first used to investigate the formation of H-type blood vessels in critical femoral defects and promotion of bone repair. Our results showed that the application of DFO-GMs hydrogel complex expanded the distribution range and expression time of H-type vessels, and was positively correlated with the number of osteoblasts and volume of new bone tissue. These results expanded the local application approach of DFO and provide a theoretical basis for targeting H-type vessels to treat large femoral defects.
Collapse
Affiliation(s)
- Yuwei Zeng
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Chuang Huang
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Dongming Duan
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Aiju Lou
- Department of Rheumatology, Liwan Central Hospital of Guangzhou, 35 Liwan Road, Guangzhou 510030, China
| | - Yuan Guo
- Department of Stomatology, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Tianhua Xiao
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Jianguo Wei
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Song Liu
- Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Zhao Wang
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Qihao Yang
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Lei Zhou
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China.
| | - Zenghui Wu
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China.
| | - Le Wang
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China.
| |
Collapse
|
35
|
Endothelial PDGF-BB/PDGFR-β signaling promotes osteoarthritis by enhancing angiogenesis-dependent abnormal subchondral bone formation. Bone Res 2022; 10:58. [PMID: 36031625 PMCID: PMC9420732 DOI: 10.1038/s41413-022-00229-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 06/14/2022] [Accepted: 07/06/2022] [Indexed: 12/11/2022] Open
Abstract
The mechanisms that coordinate the shift from joint homeostasis to osteoarthritis (OA) remain unknown. No pharmacological intervention can currently prevent the progression of osteoarthritis. Accumulating evidence has shown that subchondral bone deterioration is a primary trigger for overlying cartilage degeneration. We previously found that H-type vessels modulate aberrant subchondral bone formation during the pathogenesis of OA. However, the mechanism responsible for the elevation of H-type vessels in OA is still unclear. Here, we found that PDGFR-β expression, predominantly in the CD31hiEmcnhi endothelium, was substantially elevated in subchondral bones from OA patients and rodent OA models. A mouse model of OA with deletion of PDGFR-β in endothelial cells (ECs) exhibited fewer H-type vessels, ameliorated subchondral bone deterioration and alleviated overlying cartilage degeneration. Endothelial PDGFR-β promotes angiogenesis through the formation of the PDGFR-β/talin1/FAK complex. Notably, endothelium-specific inhibition of PDGFR-β by local injection of AAV9 in subchondral bone effectively attenuated the pathogenesis of OA compared with that of the vehicle-treated controls. Based on the results from this study, targeting PDGFR-β is a novel and promising approach for the prevention or early treatment of OA.
Collapse
|
36
|
Jacob L, de Brito Neto J, Lenck S, Corcy C, Benbelkacem F, Geraldo LH, Xu Y, Thomas JM, El Kamouh MR, Spajer M, Potier MC, Haik S, Kalamarides M, Stankoff B, Lehericy S, Eichmann A, Thomas JL. Conserved meningeal lymphatic drainage circuits in mice and humans. J Exp Med 2022; 219:e20220035. [PMID: 35776089 PMCID: PMC9253621 DOI: 10.1084/jem.20220035] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/07/2022] [Accepted: 06/08/2022] [Indexed: 12/19/2022] Open
Abstract
Meningeal lymphatic vessels (MLVs) were identified in the dorsal and caudobasal regions of the dura mater, where they ensure waste product elimination and immune surveillance of brain tissues. Whether MLVs exist in the anterior part of the murine and human skull and how they connect with the glymphatic system and extracranial lymphatics remained unclear. Here, we used light-sheet fluorescence microscopy (LSFM) imaging of mouse whole-head preparations after OVA-A555 tracer injection into the cerebrospinal fluid (CSF) and performed real-time vessel-wall (VW) magnetic resonance imaging (VW-MRI) after systemic injection of gadobutrol in patients with neurological pathologies. We observed a conserved three-dimensional anatomy of MLVs in mice and humans that aligned with dural venous sinuses but not with nasal CSF outflow, and we discovered an extended anterior MLV network around the cavernous sinus, with exit routes through the foramina of emissary veins. VW-MRI may provide a diagnostic tool for patients with CSF drainage defects and neurological diseases.
Collapse
Affiliation(s)
- Laurent Jacob
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
- Paris Cardiovascular Research Center, Institut National de la Santé et de la Recherche Médicale, Université de Paris, Paris, France
| | - Jose de Brito Neto
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
- Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Stephanie Lenck
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
- Department of Neuroradiology, Pitie-Salpêtrière Hospital, Sorbonne University, Paris, France
| | - Celine Corcy
- Department of Neuroradiology, Pitie-Salpêtrière Hospital, Sorbonne University, Paris, France
| | | | - Luiz Henrique Geraldo
- Paris Cardiovascular Research Center, Institut National de la Santé et de la Recherche Médicale, Université de Paris, Paris, France
- Department of Internal Medicine, Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT
| | - Yunling Xu
- Paris Cardiovascular Research Center, Institut National de la Santé et de la Recherche Médicale, Université de Paris, Paris, France
| | - Jean-Mickael Thomas
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
| | - Marie-Renee El Kamouh
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
| | - Myriam Spajer
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
| | - Marie-Claude Potier
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
| | - Stephane Haik
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
| | - Michel Kalamarides
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
- Department of Neurosurgery, Pitie-Salpêtrière Hospital, Sorbonne University, Paris, France
| | - Bruno Stankoff
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
- Department of Neurology, St Antoine Hospital, Assistance Publique Hôpitaux de Paris – Sorbonne, Paris, France
| | - Stephane Lehericy
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
- Department of Neuroradiology, Pitie-Salpêtrière Hospital, Sorbonne University, Paris, France
- Centre for NeuroImaging Research, Institut du Cerveau et de la Moelle épinière, Paris, France
| | - Anne Eichmann
- Paris Cardiovascular Research Center, Institut National de la Santé et de la Recherche Médicale, Université de Paris, Paris, France
- Department of Internal Medicine, Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT
| | - Jean-Leon Thomas
- Institut du Cerveau, Pitié-Salpêtrière Hospital, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France
- Department of Neurology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
37
|
Roth DM, Souter K, Graf D. Craniofacial sutures: Signaling centres integrating mechanosensation, cell signaling, and cell differentiation. Eur J Cell Biol 2022; 101:151258. [PMID: 35908436 DOI: 10.1016/j.ejcb.2022.151258] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 11/03/2022] Open
Abstract
Cranial sutures are dynamic structures in which stem cell biology, bone formation, and mechanical forces interface, influencing the shape of the skull throughout development and beyond. Over the past decade, there has been significant progress in understanding mesenchymal stromal cell (MSC) differentiation in the context of suture development and genetic control of suture pathologies, such as craniosynostosis. More recently, the mechanosensory function of sutures and the influence of mechanical signals on craniofacial development have come to the forefront. There is currently a gap in understanding of how mechanical signals integrate with MSC differentiation and ossification to ensure appropriate bone development and mediate postnatal growth surrounding sutures. In this review, we discuss the role of mechanosensation in the context of cranial sutures, and how mechanical stimuli are converted to biochemical signals influencing bone growth, suture patency, and fusion through mediation of cell differentiation. We integrate key knowledge from other paradigms where mechanosensation forms a critical component, such as bone remodeling and orthodontic tooth movement. The current state of the field regarding genetic, cellular, and physiological mechanisms of mechanotransduction will be contextualized within suture biology.
Collapse
Affiliation(s)
- Daniela Marta Roth
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| | - Katherine Souter
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| | - Daniel Graf
- School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada; Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
38
|
Shi H, Zhao Z, Jiang W, Zhu P, Zhou N, Huang X. A Review Into the Insights of the Role of Endothelial Progenitor Cells on Bone Biology. Front Cell Dev Biol 2022; 10:878697. [PMID: 35686054 PMCID: PMC9173585 DOI: 10.3389/fcell.2022.878697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/11/2022] [Indexed: 11/23/2022] Open
Abstract
In addition to its important transport functions, the skeletal system is involved in complex biological activities for the regulation of blood vessels. Endothelial progenitor cells (EPCs), as stem cells of endothelial cells (ECs), possess an effective proliferative capacity and a powerful angiogenic capacity prior to their differentiation. They demonstrate synergistic effects to promote bone regeneration and vascularization more effectively by co-culturing with multiple cells. EPCs demonstrate a significant therapeutic potential for the treatment of various bone diseases by secreting a combination of growth factors, regulating cellular functions, and promoting bone regeneration. In this review, we retrospect the definition and properties of EPCs, their interaction with mesenchymal stem cells, ECs, smooth muscle cells, and immune cells in bone regeneration, vascularization, and immunity, summarizing their mechanism of action and contribution to bone biology. Additionally, we generalized their role and potential mechanisms in the treatment of various bone diseases, possibly indicating their clinical application.
Collapse
Affiliation(s)
- Henglei Shi
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Disease Treatment, Guangxi Clinical Research Center for Craniofacia Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surg Deformity, Nanning, China
| | - Zhenchen Zhao
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Disease Treatment, Guangxi Clinical Research Center for Craniofacia Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surg Deformity, Nanning, China
| | - Weidong Jiang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Disease Treatment, Guangxi Clinical Research Center for Craniofacia Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surg Deformity, Nanning, China
| | - Peiqi Zhu
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Disease Treatment, Guangxi Clinical Research Center for Craniofacia Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surg Deformity, Nanning, China
| | - Nuo Zhou
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Disease Treatment, Guangxi Clinical Research Center for Craniofacia Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surg Deformity, Nanning, China
| | - Xuanping Huang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Disease Treatment, Guangxi Clinical Research Center for Craniofacia Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surg Deformity, Nanning, China
| |
Collapse
|
39
|
Buckley MW, McGavern DB. Immune dynamics in the CNS and its barriers during homeostasis and disease. Immunol Rev 2022; 306:58-75. [PMID: 35067941 PMCID: PMC8852772 DOI: 10.1111/imr.13066] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/30/2021] [Accepted: 01/03/2022] [Indexed: 12/11/2022]
Abstract
The central nervous system (CNS) has historically been viewed as an immunologically privileged site, but recent studies have uncovered a vast landscape of immune cells that reside primarily along its borders. While microglia are largely responsible for surveying the parenchyma, CNS barrier sites are inhabited by a plethora of different innate and adaptive immune cells that participate in everything from the defense against microbes to the maintenance of neural function. Static and dynamic imaging studies have revolutionized the field of neuroimmunology by providing detailed maps of CNS immune cells as well as information about how these cells move, organize, and interact during steady-state and inflammatory conditions. These studies have also redefined our understanding of neural-immune interactions at a cellular level and reshaped our conceptual view of immune privilege in this specialized compartment. This review will focus on insights gained using imaging techniques in the field of neuroimmunology, with an emphasis on anatomy and CNS immune dynamics during homeostasis, infectious diseases, injuries, and aging.
Collapse
Affiliation(s)
- Monica W. Buckley
- Viral Immunology and Intravital Imaging Section National Institute of Neurological Disorders and Stroke National Institutes of Health Bethesda Maryland USA
| | - Dorian B. McGavern
- Viral Immunology and Intravital Imaging Section National Institute of Neurological Disorders and Stroke National Institutes of Health Bethesda Maryland USA
| |
Collapse
|
40
|
Rindone AN, Grayson WL. Illuminating the Regenerative Microenvironment: Emerging Quantitative Imaging Technologies for Craniofacial Bone Tissue Engineering. ACS Biomater Sci Eng 2022; 8:4610-4612. [PMID: 35157425 DOI: 10.1021/acsbiomaterials.1c01373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Tissue engineering has the potential to revolutionize treatments for patients suffering from critical-sized craniofacial bone defects, but it has yet to make a substantial impact in clinical practice. One of the barriers to improving the design of tissue-engineered bone grafts (TEBGs) is the lack of adequate techniques to study how transplanted cells, host cells, and biomaterials interact to facilitate the dynamic healing process. In this perspective, we discuss recent advances in quantitative imaging that may be adapted to provide high spatiotemporal resolution of the 3D tissue microenvironment during cranial bone regeneration. The adoption and application of these imaging technologies will provide a more rigorous framework for evaluating TEBG performance and enable the development of next-generation TEBGs for craniofacial repair.
Collapse
Affiliation(s)
- Alexandra N Rindone
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States.,Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Warren L Grayson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States.,Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland 21205 United States.,Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland 21205, United States.,Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21205, United States
| |
Collapse
|
41
|
Rosenblum JS, Cappadona AJ, Lookian PP, Chandrashekhar V, Bryant JP, Chandrashekhar V, Zhao DY, Knutsen RH, Donahue DR, McGavern DB, Kozel BA, Heiss JD, Pacak K, Zhuang Z. Non-invasive in situ Visualization of the Murine Cranial Vasculature. CELL REPORTS METHODS 2022; 2:100151. [PMID: 35373177 PMCID: PMC8967186 DOI: 10.1016/j.crmeth.2021.100151] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 09/29/2021] [Accepted: 12/20/2021] [Indexed: 11/16/2022]
Abstract
Understanding physiologic and pathologic central nervous system function depends on our ability to map the entire in situ cranial vasculature and neurovascular interfaces. To accomplish this, we developed a non-invasive workflow to visualize murine cranial vasculature via polymer casting of vessels, iterative sample processing and micro-computed tomography, and automatic deformable image registration, feature extraction, and visualization. This methodology is applicable to any tissue and allows rapid exploration of normal and altered pathologic states.
Collapse
Affiliation(s)
| | - Anthony J. Cappadona
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Pashayar P. Lookian
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Jean-Paul Bryant
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - David Y. Zhao
- Department of Neurosurgery, Medstar Georgetown University Hospital, Washington, DC 20007, USA
| | - Russell H. Knutsen
- Laboratory of Vascular and Matrix Genetics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Danielle R. Donahue
- Mouse Imaging Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dorian B. McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Beth A. Kozel
- Laboratory of Vascular and Matrix Genetics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - John D. Heiss
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Karel Pacak
- Eunice Kennedy Shriver National Institute of Child Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zhengping Zhuang
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|