1
|
Mizoguchi T. In vivo dynamics of hard tissue-forming cell origins: Insights from Cre/loxP-based cell lineage tracing studies. JAPANESE DENTAL SCIENCE REVIEW 2024; 60:109-119. [PMID: 38406212 PMCID: PMC10885318 DOI: 10.1016/j.jdsr.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/27/2024] Open
Abstract
Bone tissue provides structural support for our bodies, with the inner bone marrow (BM) acting as a hematopoietic organ. Within the BM tissue, two types of stem cells play crucial roles: mesenchymal stem cells (MSCs) (or skeletal stem cells) and hematopoietic stem cells (HSCs). These stem cells are intricately connected, where BM-MSCs give rise to bone-forming osteoblasts and serve as essential components in the BM microenvironment for sustaining HSCs. Despite the mid-20th century proposal of BM-MSCs, their in vivo identification remained elusive owing to a lack of tools for analyzing stemness, specifically self-renewal and multipotency. To address this challenge, Cre/loxP-based cell lineage tracing analyses are being employed. This technology facilitated the in vivo labeling of specific cells, enabling the tracking of their lineage, determining their stemness, and providing a deeper understanding of the in vivo dynamics governing stem cell populations responsible for maintaining hard tissues. This review delves into cell lineage tracing studies conducted using commonly employed genetically modified mice expressing Cre under the influence of LepR, Gli1, and Axin2 genes. These studies focus on research fields spanning long bones and oral/maxillofacial hard tissues, offering insights into the in vivo dynamics of stem cell populations crucial for hard tissue homeostasis.
Collapse
|
2
|
Wei J, Dong R, Ma Y, Wang J, Tian S, Tu X, Mu Z, Liu YQ. Single-cell sequencing reveals that specnuezhenide protects against osteoporosis via activation of METTL3 in LEPR + BMSCs. Eur J Pharmacol 2024; 981:176908. [PMID: 39154827 DOI: 10.1016/j.ejphar.2024.176908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND Osteoporosis (OP) has garnered significant attention due to its substantial morbidity and mortality rates, imposing considerable health burdens on societies worldwide. However, the molecular mechanisms underlying osteoporosis pathogenesis remain largely elusive, and the available therapeutic interventions are limited. Therefore, there is an urgent need for innovative strategies in the treatment of osteoporosis. PURPOSE The primary objective of this study was to elucidate the molecular mechanisms underlying osteoporosis pathogenesis using single-cell RNA sequencing (scRNA-seq), thereby proposing novel therapeutic agents. METHODS The mice osteoporosis model was established through bilateral ovariectomy. Micro-computed tomography (μCT) and hematoxylin and eosin (H&E) staining were employed to assess the pathogenesis of osteoporosis. scRNA-seq was utilized to identify and analyze distinct molecular mechanisms and sub-clusters. Gradient dilution analysis was used to obtain specific sub-clusters, which were further validated by immunofluorescence staining and flow cytometry analysis. Molecular docking and cellular thermal shift assay (CETSA) were applied for screening potential agents in the TCMSPs database. Alkaline phosphatase (ALP) activity and alizarin red S (ARS) staining were performed to evaluate the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). Osteogenic organoids analysis was employed to assess the proliferation and sphere-forming ability of BMSCs. Quantitative real-time PCR (qRT-PCR) and western blot analysis were conducted to investigate signaling pathways. Wound healing assay and tube formation analysis were employed to evaluate the angiogenesis of endothelial cells. RESULTS The scRNA-seq analysis revealed the crucial role of LEPR+ BMSCs in the pathogenesis of osteoporosis, which was confirmed by immunofluorescence staining of the epiphysis. Subsequently, the LEPR+ BMSCs were obtained by gradient dilution analysis and identified by immunofluorescence staining and flow cytometry. Accordingly, specnuezhenide (Spe) was screened and identified as a potential compound targeting METTL3 from the TCMSPs database. Spe promoted bone formation as evidenced by μ-CT, and H&E analysis. Additionally, Spe enhanced the osteogenic capacity of LEPR+ BMSCs through ALP and ARS assay. Notably, METTL3 pharmacological inhibitors S-Adenosylhomocysteine (SAH) attenuated the aforementioned osteo-protective effects of Spe. Particularly, Spe enhanced the LEPR+ BMSCs-dependent angiogenesis through the secretion of SLIT3, which was abolished by SAH in LEPR+ BMSCs. CONCLUSION Collectively, these findings suggest that Spe could enhance the osteogenic potential of LEPR+ BMSCs and promote LEPR+ BMSCs-dependent angiogenesis by activating METTL3 in LEPR+ BMSCs, indicating its potential as an ideal therapeutic agent for clinical treatment of osteoporosis.
Collapse
Affiliation(s)
- Jun Wei
- Shandong University of Traditional Chinese Medicine, Jinan, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Renchao Dong
- Shandong University of Traditional Chinese Medicine, Jinan, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yu Ma
- Shandong University of Traditional Chinese Medicine, Jinan, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jie Wang
- Shandong University of Traditional Chinese Medicine, Jinan, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shuo Tian
- Shandong University of Traditional Chinese Medicine, Jinan, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinyi Tu
- Shandong University of Traditional Chinese Medicine, Jinan, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhenqiang Mu
- Chongqing Key Laboratory of High Active Traditional Chinese Medicine Delivery System & Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing, China.
| | - Yan-Qiu Liu
- Shandong University of Traditional Chinese Medicine, Jinan, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
3
|
Melis S, Trompet D, Chagin AS, Maes C. Skeletal stem and progenitor cells in bone physiology, ageing and disease. Nat Rev Endocrinol 2024:10.1038/s41574-024-01039-y. [PMID: 39379711 DOI: 10.1038/s41574-024-01039-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/30/2024] [Indexed: 10/10/2024]
Abstract
Skeletal stem cells (SSCs) and related progenitors with osteogenic potential, collectively termed skeletal stem and/or progenitor cells (SSPCs), are crucial for providing osteoblasts for bone formation during homeostatic tissue turnover and fracture repair. Besides mediating normal bone physiology, they also have important roles in various metabolic bone diseases, including osteoporosis. SSPCs are of tremendous interest because they represent prime future targets for osteoanabolic therapies and bone regenerative medicine. Remarkable progress has been made in characterizing various SSC and SSPC populations in postnatal bone. SSPCs exist in the periosteum and within the bone marrow stroma, including subsets localizing around arteriolar and sinusoidal blood vessels; they can display osteogenic, chondrogenic, adipogenic and/or fibroblastic potential, and exert critical haematopoiesis-supportive functions. However, much remains to be clarified. By the current markers, bona fide SSCs are commonly contained within broader SSPC populations characterized by considerable heterogeneity and overlap, whose common versus specific functions in health and disease have not been fully unravelled. Here, we review the present knowledge of the identity, fates and relationships of SSPC populations in the postnatal bone environment, their contributions to bone maintenance, the changes observed upon ageing, and the effect of metabolic diseases such as osteoporosis and diabetes mellitus.
Collapse
Affiliation(s)
- Seppe Melis
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Dana Trompet
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Andrei S Chagin
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Christa Maes
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Leuven, Belgium.
| |
Collapse
|
4
|
Li Z, Lin J, Wu J, Suo J, Wang Z. The Hippo signalling pathway in bone homeostasis: Under the regulation of mechanics and aging. Cell Prolif 2024; 57:e13652. [PMID: 38700015 PMCID: PMC11471399 DOI: 10.1111/cpr.13652] [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: 02/01/2024] [Revised: 04/04/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024] Open
Abstract
The Hippo signalling pathway is a conserved kinase cascade that orchestrates diverse cellular processes, such as proliferation, apoptosis, lineage commitment and stemness. With the onset of society ages, research on skeletal aging-mechanics-bone homeostasis has exploded. In recent years, aging and mechanical force in the skeletal system have gained groundbreaking research progress. Under the regulation of mechanics and aging, the Hippo signalling pathway has a crucial role in the development and homeostasis of bone. We synthesize the current knowledge on the role of the Hippo signalling pathway, particularly its downstream effectors yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ), in bone homeostasis. We discuss the regulation of the lineage specification and function of different skeletal cell types by the Hippo signalling pathway. The interactions of the Hippo signalling pathway with other pathways, such as Wnt, transforming growth factor beta and nuclear factor kappa-B, are also mentioned because of their importance for modulating bone homeostasis. Furthermore, YAP/TAZ have been extensively studied as mechanotransducers. Due to space limitations, we focus on reviewing how mechanical forces and aging influence cell fate, communications and homeostasis through a dysregulated Hippo signalling pathway.
Collapse
Affiliation(s)
- Zhengda Li
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences and Shanghai Jing'an District Central HospitalFudan UniversityShanghaiChina
| | - Junqing Lin
- Institute of Microsurgery on Extremities, and Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine ShanghaiShanghaiChina
| | - Jing Wu
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences and Shanghai Jing'an District Central HospitalFudan UniversityShanghaiChina
| | - Jinlong Suo
- Institute of Microsurgery on Extremities, and Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine ShanghaiShanghaiChina
| | - Zuoyun Wang
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences and Shanghai Jing'an District Central HospitalFudan UniversityShanghaiChina
| |
Collapse
|
5
|
Poonia N, Jadhav NV, Mamatha D, Garg M, Kabra A, Bhatia A, Ojha S, Lather V, Pandita D. Nanotechnology-assisted combination drug delivery: a progressive approach for the treatment of acute myeloid leukemia. Ther Deliv 2024; 15:893-910. [PMID: 39268925 PMCID: PMC11497954 DOI: 10.1080/20415990.2024.2394012] [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: 01/11/2024] [Accepted: 08/15/2024] [Indexed: 09/15/2024] Open
Abstract
Acute myeloid leukemia (AML), a heterogeneous hematopoietic cancer prevalent in adults, has been a leading cause of leukemia-associated deaths for decades. Despite advancements in understanding its pathology and pharmacological targets, therapeutic strategies have seen minimal change. The standard treatment, combining cytarabine and anthracycline, has persisted, accompanied by challenges such as pharmacokinetic issues and non-specific drug delivery, leading to severe side effects. Nanotechnology offers a promising solution through combination drug delivery. FDA-approved CPX351 (VYXEOS™) a liposomal formulation delivering doxorubicin and cytarabine, exemplifies enhanced therapeutic efficacy. Ongoing research explores various nanocarriers for delivering multiple bioactives, addressing drug targeting, pharmacokinetics and chemoresistance. This review highlights nanotechnology-based combination therapies for the effective management of AML, presenting a potential breakthrough in leukemia.
Collapse
Affiliation(s)
- Neelam Poonia
- University Institute of Pharma Sciences, Chandigarh University, Gharuan, Mohali, Punjab, India
| | - Nikita Vijay Jadhav
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences & Research (DIPSAR), Delhi Pharmaceutical Sciences & Research University, Pushp Vihar, Govt. of NCT of Delhi, New Delhi, 110017, India
| | - Davuluri Mamatha
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences & Research (DIPSAR), Delhi Pharmaceutical Sciences & Research University, Pushp Vihar, Govt. of NCT of Delhi, New Delhi, 110017, India
| | - Manoj Garg
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector-125, Noida, 201313, India
| | - Atul Kabra
- University Institute of Pharma Sciences, Chandigarh University, Gharuan, Mohali, Punjab, India
| | - Amit Bhatia
- Department of Pharmaceutical Sciences & Technology, Maharaja Ranjit Singh Punjab Technical University (Govt of Punjab), Dabwali Road, Bathinda, Punjab, 151001, India
| | - Shreesh Ojha
- Pharmacology, College of Medicine & Health Sciences, P.O. Box 15551, Al Ain, UAE
| | - Viney Lather
- Amity Institute of Pharmacy, Amity University, Noida, 201313, Uttar Pradesh, India
| | - Deepti Pandita
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences & Research (DIPSAR), Delhi Pharmaceutical Sciences & Research University, Pushp Vihar, Govt. of NCT of Delhi, New Delhi, 110017, India
- Centre for Advanced Formulation & Technology (CAFT), Delhi Pharmaceutical Sciences & Research University, Pushp Vihar, Govt. of NCT of Delhi, New Delhi, 110017, India
| |
Collapse
|
6
|
Cain TL, Derecka M, McKinney-Freeman S. The role of the haematopoietic stem cell niche in development and ageing. Nat Rev Mol Cell Biol 2024:10.1038/s41580-024-00770-8. [PMID: 39256623 DOI: 10.1038/s41580-024-00770-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2024] [Indexed: 09/12/2024]
Abstract
Blood production depends on rare haematopoietic stem cells (HSCs) and haematopoietic stem and progenitor cells (HSPCs) that ultimately take up residence in the bone marrow during development. HSPCs and HSCs are subject to extrinsic regulation by the bone marrow microenvironment, or niche. Studying the interactions between HSCs and their niche is critical for improving ex vivo culturing conditions and genetic manipulation of HSCs, which is pivotal for improving autologous HSC therapies and transplantations. Additionally, understanding how the complex molecular network in the bone marrow is altered during ageing is paramount for developing novel therapeutics for ageing-related haematopoietic disorders. HSCs are unique amongst stem and progenitor cell pools in that they engage with multiple physically distinct niches during their ontogeny. HSCs are specified from haemogenic endothelium in the aorta, migrate to the fetal liver and, ultimately, colonize their final niche in the bone marrow. Recent studies employing single-cell transcriptomics and microscopy have identified novel cellular interactions that govern HSC specification and engagement with their niches throughout ontogeny. New lineage-tracing models and microscopy tools have raised questions about the numbers of HSCs specified, as well as the functional consequences of HSCs interacting with each developmental niche. Advances have also been made in understanding how these niches are modified and perturbed during ageing, and the role of these altered interactions in haematopoietic diseases. In this Review, we discuss these new findings and highlight the questions that remain to be explored.
Collapse
Affiliation(s)
- Terri L Cain
- Department of Haematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Marta Derecka
- Department of Haematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | |
Collapse
|
7
|
Lee K, Dissanayake W, MacLiesh M, Hong CL, Yin Z, Kawano Y, Kaszuba CM, Kawano H, Quarato ER, Marples B, Becker M, Bajaj J, Calvi LM, Yeh SCA. Ultralow-dose irradiation enables engraftment and intravital tracking of disease initiating niches in clonal hematopoiesis. Sci Rep 2024; 14:20486. [PMID: 39227700 PMCID: PMC11372138 DOI: 10.1038/s41598-024-71307-4] [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: 05/09/2024] [Accepted: 08/27/2024] [Indexed: 09/05/2024] Open
Abstract
Recent advances in imaging suggested that spatial organization of hematopoietic cells in their bone marrow microenvironment (niche) regulates cell expansion, governing progression, and leukemic transformation of hematological clonal disorders. However, our ability to interrogate the niche in pre-malignant conditions has been limited, as standard murine models of these diseases rely largely on transplantation of the mutant clones into conditioned mice where the marrow microenvironment is compromised. Here, we leveraged live-animal microscopy and ultralow dose whole body or focal irradiation to capture single cells and early expansion of benign/pre-malignant clones in the functionally preserved microenvironment. 0.5 Gy whole body irradiation (WBI) allowed steady engraftment of cells beyond 30 weeks compared to non-conditioned controls. In-vivo tracking and functional analyses of the microenvironment showed no change in vessel integrity, cell viability, and HSC-supportive functions of the stromal cells, suggesting minimal inflammation after the radiation insult. The approach enabled in vivo imaging of Tet2+/- and its healthy counterpart, showing preferential localization within a shared microenvironment while forming discrete micro-niches. Notably, stationary association with the niche only occurred in a subset of cells and would not be identified without live imaging. This strategy may be broadly applied to study clonal disorders in a spatial context.
Collapse
Affiliation(s)
- Kevin Lee
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14623, USA
| | - Wimeth Dissanayake
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14623, USA
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - Melissa MacLiesh
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14623, USA
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - Cih-Li Hong
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14623, USA
- Department of Physiology/Pharmacology, University of Rochester Medical Center, Rochester, NY, USA
| | - Zi Yin
- Institute of Optics, University of Rochester, Rochester, NY, USA
| | - Yuko Kawano
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Christina M Kaszuba
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Hiroki Kawano
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Emily R Quarato
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Department of Toxicology, University of Rochester Medical Center, Rochester, NY, USA
| | - Brian Marples
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY, USA
| | - Michael Becker
- Department of Medicine, Indiana University, Indianapolis, IN, USA
| | - Jeevisha Bajaj
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
| | - Laura M Calvi
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14623, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Shu-Chi A Yeh
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA.
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14623, USA.
- Department of Physiology/Pharmacology, University of Rochester Medical Center, Rochester, NY, USA.
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
8
|
Wu R, Wu J, Jin H, Ma H, Huang H, Xu W, Sun S, Liu X, Dong K, Xie Y, Zeng J, Wang F. Olink and gut microbial metabolomics reveal new biomarkers for the prediction and diagnosis of PMOP. J Bone Miner Metab 2024; 42:503-515. [PMID: 39153113 DOI: 10.1007/s00774-024-01545-z] [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: 03/19/2024] [Accepted: 08/02/2024] [Indexed: 08/19/2024]
Abstract
LNTRODUCTION Postmenopausal osteoporosis (PMOP) can cause postmenopausal women to experience pain and interference. Identifying and exploring potential early diagnostic biomarkers of PMOP is of substantial clinical value and social significance. This study aimed to screen for potential novel diagnostic biomarkers of PMOP through a multiomics approach, providing new directions and ideas for the early prevention and treatment of this disease. MATERIALS AND METHODS Fifteen postmenopausal women with osteoporosis and 12 without were recruited. Clinical information was collected, and various clinical biochemical parameters were tested. Plasma and fecal samples were collected and analyzed using Olink proteomics and gut microbial metabolomics. RESULTS The functions of the differentially abundant metabolites were mainly related to autophagy and arginine and proline metabolism and were involved in immunoinflammatory metabolic processes. Olink showed significant differences in the expression of seven inflammation-related proteins between the two groups. CONCLUSION We demonstrated that metabolic differences between PMOP patients and healthy controls were associated with inflammatory responses and found seven proteins with significant differences. Among these proteins, CDCP1, IL10, and IL-1alpha combined with clinical indicators had high discriminant efficiency in identifying PMOP. This is also the first study to demonstrate noteworthy changes in CDCP1 levels in patients with PMOP.
Collapse
Affiliation(s)
- Ruizhe Wu
- Department of Orthopaedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Jie Wu
- Department of Orthopaedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Hui Jin
- Department of Orthopaedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Huaiyu Ma
- Department of Orthopaedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Hongxing Huang
- Department of Orthopaedics, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Wuji Xu
- Department of Orthopaedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Shaoqiu Sun
- Department of Orthopaedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Xiaolan Liu
- Department of Orthopaedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Kefang Dong
- Department of Orthopaedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Yisong Xie
- Department of Orthopaedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Jingqi Zeng
- Department of Orthopaedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China.
| | - Fan Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China.
| |
Collapse
|
9
|
Li N, Shi B, Li Z, Han J, Sun J, Huang H, Yallowitz AR, Bok S, Xiao S, Wu Z, Chen Y, Xu Y, Qin T, Huang R, Zheng H, Shen R, Meng L, Greenblatt MB, Xu R. Schnurri-3 inhibition rescues skeletal fragility and vascular skeletal stem cell niche pathology in the OIM model of osteogenesis imperfecta. Bone Res 2024; 12:46. [PMID: 39183236 PMCID: PMC11345453 DOI: 10.1038/s41413-024-00349-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 06/18/2024] [Accepted: 06/24/2024] [Indexed: 08/27/2024] Open
Abstract
Osteogenesis imperfecta (OI) is a disorder of low bone mass and increased fracture risk due to a range of genetic variants that prominently include mutations in genes encoding type I collagen. While it is well known that OI reflects defects in the activity of bone-forming osteoblasts, it is currently unclear whether OI also reflects defects in the many other cell types comprising bone, including defects in skeletal vascular endothelium or the skeletal stem cell populations that give rise to osteoblasts and whether correcting these broader defects could have therapeutic utility. Here, we find that numbers of skeletal stem cells (SSCs) and skeletal arterial endothelial cells (AECs) are augmented in Col1a2oim/oim mice, a well-studied animal model of moderate to severe OI, suggesting that disruption of a vascular SSC niche is a feature of OI pathogenesis. Moreover, crossing Col1a2oim/oim mice to mice lacking a negative regulator of skeletal angiogenesis and bone formation, Schnurri 3 (SHN3), not only corrected the SSC and AEC phenotypes but moreover robustly corrected the bone mass and spontaneous fracture phenotypes. As this finding suggested a strong therapeutic utility of SHN3 inhibition for the treatment of OI, a bone-targeting AAV was used to mediate Shn3 knockdown, rescuing the Col1a2oim/oim phenotype and providing therapeutic proof-of-concept for targeting SHN3 for the treatment of OI. Overall, this work both provides proof-of-concept for inhibition of the SHN3 pathway and more broadly addressing defects in the stem/osteoprogenitor niche as is a strategy to treat OI.
Collapse
Affiliation(s)
- Na Li
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Baohong Shi
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Zan Li
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Jie Han
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Jun Sun
- Research Division, Hospital for Special Surgery, New York, NY, 10065, USA
| | - Haitao Huang
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Alisha R Yallowitz
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Seoyeon Bok
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Shuang Xiao
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Zuoxing Wu
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Yu Chen
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Yan Xu
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Tian Qin
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Rui Huang
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Haiping Zheng
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Rong Shen
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Lin Meng
- Department of Electronic and Computer Engineering, Ritsumeikan University, Kusatsu, Shiga, 525-8577, Japan
| | - Matthew B Greenblatt
- Research Division, Hospital for Special Surgery, New York, NY, 10065, USA.
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, 10065, USA.
| | - Ren Xu
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China.
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, 361102, China.
| |
Collapse
|
10
|
Luo Z, Wei W, Qiu D, Su Z, Liu L, Zhou H, Cui H, Yang L. Rejuvenation of BMSCs senescence by pharmacological enhancement of TFEB-mediated autophagy alleviates aged-related bone loss and extends lifespan in middle aged mice. Bone Res 2024; 12:45. [PMID: 39164234 PMCID: PMC11336217 DOI: 10.1038/s41413-024-00351-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 06/30/2024] [Accepted: 07/12/2024] [Indexed: 08/22/2024] Open
Abstract
Bone marrow stromal/stem cells (BMSCs) are generally considered as common progenitors for both osteoblasts and adipocytes in the bone marrow, but show preferential differentiation into adipocytes rather than osteoblasts under aging, thus leading to senile osteoporosis. Accumulated evidences indicate that rejuvenation of BMSCs by autophagic enhancement delays bone aging. Here we synthetized and demonstrated a novel autophagy activator, CXM102 that could induce autophagy in aged BMSCs, resulting in rejuvenation and preferential differentiation into osteoblasts of BMSCs. Furthermore, CXM102 significantly stimulated bone anabolism, reduced marrow adipocytes, and delayed bone loss in middle-age male mice. Mechanistically, CXM102 promoted transcription factor EB (TFEB) nuclear translocation and favored osteoblasts formation both in vitro and in vivo. Moreover, CXM102 decreased serum levels of inflammation and reduced organ fibrosis, leading to a prolonger lifespan in male mice. Our results indicated that CXM102 could be used as an autophagy inducer to rejuvenate BMSCs and shed new lights on strategies for senile osteoporosis and healthyspan improvement.
Collapse
Affiliation(s)
- Ziwei Luo
- College of Orthopedics, Guangxi University of Chinese Medicine, Nanning, 530200, Guangxi, China.
| | - Wanyi Wei
- Faculty of Chinese Medicine Science, Guangxi University of Chinese Medicine, Nanning, 530200, Guangxi, China
| | - Dawei Qiu
- Department of Physical Education, Guangxi University of Chinese Medicine, Nanning, 530200, Guangxi, China
| | - Zixia Su
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, 530200, Guangxi, China
| | - Liangpu Liu
- College of Orthopedics, Guangxi University of Chinese Medicine, Nanning, 530200, Guangxi, China
| | - Honghai Zhou
- College of Orthopedics, Guangxi University of Chinese Medicine, Nanning, 530200, Guangxi, China
| | - Hao Cui
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530200, Guangxi, China.
| | - Li Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing, 400030, China
| |
Collapse
|
11
|
Xu K, Huang RQ, Wen R, Yang Y, Cheng Y, Chang B. The role of Clec11a in bone construction and remodeling. Front Endocrinol (Lausanne) 2024; 15:1429567. [PMID: 39188913 PMCID: PMC11345164 DOI: 10.3389/fendo.2024.1429567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/25/2024] [Indexed: 08/28/2024] Open
Abstract
Bone is a dynamically active tissue whose health status is closely related to its construction and remodeling, and imbalances in bone homeostasis lead to a wide range of bone diseases. The sulfated glycoprotein C-type lectin structural domain family 11 member A (Clec11a) is a key factor in bone mass regulation that significantly promotes the osteogenic differentiation of bone marrow mesenchymal stem cells and osteoblasts and stimulates chondrocyte proliferation, thereby promoting longitudinal bone growth. More importantly, Clec11a has high therapeutic potential for treating various bone diseases and can enhance the therapeutic effects of the parathyroid hormone against osteoporosis. Clec11a is also involved in the stress/adaptive response of bone to exercise via mechanical stimulation of the cation channel Pieoz1. Clec11a plays an important role in promoting bone health and preventing bone disease and may represent a new target and novel drug for bone disease treatment. Therefore, this review aims to explore the role and possible mechanisms of Clec11a in the skeletal system, evaluate its value as a potential therapeutic target against bone diseases, and provide new ideas and strategies for basic research on Clec11a and preventing and treating bone disease.
Collapse
Affiliation(s)
- Ke Xu
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Rui-qi Huang
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Ruiming Wen
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Yao Yang
- Laboratory Management Center, Shenyang Sport University, Shenyang, Liaoning, China
| | - Yang Cheng
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Bo Chang
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China
- School of Sport Science, Zhuhai College of Science and Technology, Zhuhai, Guangdong, China
| |
Collapse
|
12
|
Pereira AL, Galli S, Nombela‐Arrieta C. Bone marrow niches for hematopoietic stem cells. Hemasphere 2024; 8:e133. [PMID: 39086665 PMCID: PMC11289431 DOI: 10.1002/hem3.133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/05/2024] [Accepted: 05/06/2024] [Indexed: 08/02/2024] Open
Abstract
Hematopoietic stem cells (HSCs) are the cornerstone of the hematopoietic system. HSCs sustain the continuous generation of mature blood derivatives while self-renewing to preserve a relatively constant pool of progenitors throughout life. Yet, long-term maintenance of functional HSCs exclusively takes place in association with their native tissue microenvironment of the bone marrow (BM). HSCs have been long proposed to reside in fixed and identifiable anatomical units found in the complex BM tissue landscape, which control their identity and fate in a deterministic manner. In the last decades, tremendous progress has been made in the dissection of the cellular and molecular fabric of the BM, the structural organization governing tissue function, and the plethora of interactions established by HSCs. Nonetheless, a holistic model of the mechanisms controlling HSC regulation in their niche is lacking to date. Here, we provide an overview of our current understanding of BM anatomy, HSC localization, and crosstalk within local cellular neighborhoods in murine and human tissues, and highlight fundamental open questions on how HSCs functionally integrate in the BM microenvironment.
Collapse
Affiliation(s)
- Ana Luísa Pereira
- Department of Medical Oncology and HematologyUniversity Hospital and University of ZurichZurichSwitzerland
| | - Serena Galli
- Department of Medical Oncology and HematologyUniversity Hospital and University of ZurichZurichSwitzerland
| | - César Nombela‐Arrieta
- Department of Medical Oncology and HematologyUniversity Hospital and University of ZurichZurichSwitzerland
| |
Collapse
|
13
|
Quarato ER, Salama NA, Calvi LM. Interplay Between Skeletal and Hematopoietic Cells in the Bone Marrow Microenvironment in Homeostasis and Aging. Curr Osteoporos Rep 2024; 22:416-432. [PMID: 38782850 DOI: 10.1007/s11914-024-00874-2] [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] [Accepted: 05/05/2024] [Indexed: 05/25/2024]
Abstract
PURPOSE OF THE REVIEW In this review, we discuss the most recent scientific advances on the reciprocal regulatory interactions between the skeletal and hematopoietic stem cell niche, focusing on immunomodulation and its interplay with the cell's mitochondrial function, and how this impacts osteoimmune health during aging and disease. RECENT FINDINGS Osteoimmunology investigates interactions between cells that make up the skeletal stem cell niche and immune system. Much work has investigated the complexity of the bone marrow microenvironment with respect to the skeletal and hematopoietic stem cells that regulate skeletal formation and immune health respectively. It has now become clear that these cellular components cooperate to maintain homeostasis and that dysfunction in their interaction can lead to aging and disease. Having a deeper, mechanistic appreciation for osteoimmune regulation will lead to better research perspective and therapeutics with the potential to improve the aging process, skeletal and hematologic regeneration, and disease targeting.
Collapse
Affiliation(s)
- Emily R Quarato
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA.
- James P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.
| | - Noah A Salama
- James P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA.
| | - Laura M Calvi
- James P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
14
|
Buenaventura T, Bagci H, Patrascan I, Graham JJ, Hipwell KD, Oldenkamp R, King JWD, Urtasun J, Young G, Mouzo D, Gomez-Cabrero D, Rowland BD, Panne D, Fisher AG, Merkenschlager M. Competition shapes the landscape of X-chromosome-linked genetic diversity. Nat Genet 2024; 56:1678-1688. [PMID: 39060501 PMCID: PMC11319201 DOI: 10.1038/s41588-024-01840-5] [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: 09/21/2023] [Accepted: 06/21/2024] [Indexed: 07/28/2024]
Abstract
X chromosome inactivation (XCI) generates clonal heterogeneity within XX individuals. Combined with sequence variation between human X chromosomes, XCI gives rise to intra-individual clonal diversity, whereby two sets of clones express mutually exclusive sequence variants present on one or the other X chromosome. Here we ask whether such clones merely co-exist or potentially interact with each other to modulate the contribution of X-linked diversity to organismal development. Focusing on X-linked coding variation in the human STAG2 gene, we show that Stag2variant clones contribute to most tissues at the expected frequencies but fail to form lymphocytes in Stag2WT Stag2variant mouse models. Unexpectedly, the absence of Stag2variant clones from the lymphoid compartment is due not solely to cell-intrinsic defects but requires continuous competition by Stag2WT clones. These findings show that interactions between epigenetically diverse clones can operate in an XX individual to shape the contribution of X-linked genetic diversity in a cell-type-specific manner.
Collapse
Affiliation(s)
- Teresa Buenaventura
- MRC LMS, Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Hakan Bagci
- MRC LMS, Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Ilinca Patrascan
- MRC LMS, Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Joshua J Graham
- Leicester Institute of Structural and Chemical Biology, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
| | - Kelsey D Hipwell
- Leicester Institute of Structural and Chemical Biology, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
| | - Roel Oldenkamp
- Division of Cell Biology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - James W D King
- MRC LMS, Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Jesus Urtasun
- MRC LMS, Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - George Young
- MRC LMS, Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Daniel Mouzo
- Translational Bioinformatics Unit, Navarrabiomed, Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - David Gomez-Cabrero
- Translational Bioinformatics Unit, Navarrabiomed, Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Bioscience Program, Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology KAUST, Thuwal, Saudi Arabia
| | - Benjamin D Rowland
- Division of Cell Biology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Daniel Panne
- Leicester Institute of Structural and Chemical Biology, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
| | - Amanda G Fisher
- MRC LMS, Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Matthias Merkenschlager
- MRC LMS, Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
15
|
Trompet D, Melis S, Chagin AS, Maes C. Skeletal stem and progenitor cells in bone development and repair. J Bone Miner Res 2024; 39:633-654. [PMID: 38696703 DOI: 10.1093/jbmr/zjae069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/11/2024] [Accepted: 04/17/2024] [Indexed: 05/04/2024]
Abstract
Bone development, growth, and repair are complex processes involving various cell types and interactions, with central roles played by skeletal stem and progenitor cells. Recent research brought new insights into the skeletal precursor populations that mediate intramembranous and endochondral bone development. Later in life, many of the cellular and molecular mechanisms determining development are reactivated upon fracture, with powerful trauma-induced signaling cues triggering a variety of postnatal skeletal stem/progenitor cells (SSPCs) residing near the bone defect. Interestingly, in this injury context, the current evidence suggests that the fates of both SSPCs and differentiated skeletal cells can be considerably flexible and dynamic, and that multiple cell sources can be activated to operate as functional progenitors generating chondrocytes and/or osteoblasts. The combined implementation of in vivo lineage tracing, cell surface marker-based cell selection, single-cell molecular analyses, and high-resolution in situ imaging has strongly improved our insights into the diversity and roles of developmental and reparative stem/progenitor subsets, while also unveiling the complexity of their dynamics, hierarchies, and relationships. Albeit incompletely understood at present, findings supporting lineage flexibility and possibly plasticity among sources of osteogenic cells challenge the classical dogma of a single primitive, self-renewing, multipotent stem cell driving bone tissue formation and regeneration from the apex of a hierarchical and strictly unidirectional differentiation tree. We here review the state of the field and the newest discoveries in the origin, identity, and fates of skeletal progenitor cells during bone development and growth, discuss the contributions of adult SSPC populations to fracture repair, and reflect on the dynamism and relationships among skeletal precursors and differentiated cell lineages. Further research directed at unraveling the heterogeneity and capacities of SSPCs, as well as the regulatory cues determining their fate and functioning, will offer vital new options for clinical translation toward compromised fracture healing and bone regenerative medicine.
Collapse
Affiliation(s)
- Dana Trompet
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, 40530 Gothenburg, Sweden
- Department of Physiology and Pharmacology, Karolinska Institute, 17177 Stockholm, Sweden
| | - Seppe Melis
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Andrei S Chagin
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, 40530 Gothenburg, Sweden
- Department of Physiology and Pharmacology, Karolinska Institute, 17177 Stockholm, Sweden
| | - Christa Maes
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
16
|
Yuan G, Lin X, Liu Y, Greenblatt MB, Xu R. Skeletal stem cells in bone development, homeostasis, and disease. Protein Cell 2024; 15:559-574. [PMID: 38442300 PMCID: PMC11259547 DOI: 10.1093/procel/pwae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/19/2024] [Indexed: 03/07/2024] Open
Abstract
Tissue-resident stem cells are essential for development and repair, and in the skeleton, this function is fulfilled by recently identified skeletal stem cells (SSCs). However, recent work has identified that SSCs are not monolithic, with long bones, craniofacial sites, and the spine being formed by distinct stem cells. Recent studies have utilized techniques such as fluorescence-activated cell sorting, lineage tracing, and single-cell sequencing to investigate the involvement of SSCs in bone development, homeostasis, and disease. These investigations have allowed researchers to map the lineage commitment trajectory of SSCs in different parts of the body and at different time points. Furthermore, recent studies have shed light on the characteristics of SSCs in both physiological and pathological conditions. This review focuses on discussing the spatiotemporal distribution of SSCs and enhancing our understanding of the diversity and plasticity of SSCs by summarizing recent discoveries.
Collapse
Affiliation(s)
- Guixin Yuan
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361005, China
- Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Xixi Lin
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361005, China
- Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Ying Liu
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361005, China
- Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Matthew B Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10065, USA
- Research Division, Hospital for Special Surgery, New York, NY 10065, USA
| | - Ren Xu
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cell, State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361005, China
- Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Organ Transplantation Institute, School of Medicine, Xiamen University, Xiamen 361102, China
| |
Collapse
|
17
|
Donnelly H, Ross E, Xiao Y, Hermantara R, Taqi AF, Doherty-Boyd WS, Cassels J, Tsimbouri PM, Dunn KM, Hay J, Cheng A, Meek RMD, Jain N, West C, Wheadon H, Michie AM, Peault B, West AG, Salmeron-Sanchez M, Dalby MJ. Bioengineered niches that recreate physiological extracellular matrix organisation to support long-term haematopoietic stem cells. Nat Commun 2024; 15:5791. [PMID: 38987295 PMCID: PMC11237034 DOI: 10.1038/s41467-024-50054-0] [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: 08/09/2022] [Accepted: 06/27/2024] [Indexed: 07/12/2024] Open
Abstract
Long-term reconstituting haematopoietic stem cells (LT-HSCs) are used to treat blood disorders via stem cell transplantation. The very low abundance of LT-HSCs and their rapid differentiation during in vitro culture hinders their clinical utility. Previous developments using stromal feeder layers, defined media cocktails, and bioengineering have enabled HSC expansion in culture, but of mostly short-term HSCs and progenitor populations at the expense of naive LT-HSCs. Here, we report the creation of a bioengineered LT-HSC maintenance niche that recreates physiological extracellular matrix organisation, using soft collagen type-I hydrogels to drive nestin expression in perivascular stromal cells (PerSCs). We demonstrate that nestin, which is expressed by HSC-supportive bone marrow stromal cells, is cytoprotective and, via regulation of metabolism, is important for HIF-1α expression in PerSCs. When CD34+ve HSCs were added to the bioengineered niches comprising nestin/HIF-1α expressing PerSCs, LT-HSC numbers were maintained with normal clonal and in vivo reconstitution potential, without media supplementation. We provide proof-of-concept that our bioengineered niches can support the survival of CRISPR edited HSCs. Successful editing of LT-HSCs ex vivo can have potential impact on the treatment of blood disorders.
Collapse
Affiliation(s)
- Hannah Donnelly
- Centre for the Cellular Microenvironment, School of Molecular Biosciences, The Advanced Research Centre, 11 Chapel Lane, University of Glasgow, Glasgow, G11 6EW, United Kingdom
| | - Ewan Ross
- Centre for the Cellular Microenvironment, School of Molecular Biosciences, The Advanced Research Centre, 11 Chapel Lane, University of Glasgow, Glasgow, G11 6EW, United Kingdom
| | - Yinbo Xiao
- Centre for the Cellular Microenvironment, School of Molecular Biosciences, The Advanced Research Centre, 11 Chapel Lane, University of Glasgow, Glasgow, G11 6EW, United Kingdom
| | - Rio Hermantara
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1QH, United Kingdom
| | - Aqeel F Taqi
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1QH, United Kingdom
| | - W Sebastian Doherty-Boyd
- Centre for the Cellular Microenvironment, School of Molecular Biosciences, The Advanced Research Centre, 11 Chapel Lane, University of Glasgow, Glasgow, G11 6EW, United Kingdom
| | - Jennifer Cassels
- School of Cancer Sciences, Paul O'Gorman Leukaemia Research Centre, Gartnavel General Hospital, University of Glasgow, Glasgow, G12 0YN, United Kingdom
| | - Penelope M Tsimbouri
- Centre for the Cellular Microenvironment, School of Molecular Biosciences, The Advanced Research Centre, 11 Chapel Lane, University of Glasgow, Glasgow, G11 6EW, United Kingdom
| | - Karen M Dunn
- School of Cancer Sciences, Paul O'Gorman Leukaemia Research Centre, Gartnavel General Hospital, University of Glasgow, Glasgow, G12 0YN, United Kingdom
| | - Jodie Hay
- School of Cancer Sciences, Paul O'Gorman Leukaemia Research Centre, Gartnavel General Hospital, University of Glasgow, Glasgow, G12 0YN, United Kingdom
| | - Annie Cheng
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, James Watt School of Engineering, The Advanced Research Centre, 11 Chapel Lane, University of Glasgow, Glasgow, G11 6EW, United Kingdom
| | - R M Dominic Meek
- Department of Trauma and Orthopaedics, Queen Elizabeth University Hospital, Glasgow, G51 4TF, United Kingdom
| | - Nikhil Jain
- Institute of Inflammation and Ageing, University of Birmingham, Queen Elizabeth Hospital, Birmingham, B15 2WB, United Kingdom
| | - Christopher West
- MRC Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh, EH16 4UU, United Kingdom
| | - Helen Wheadon
- School of Cancer Sciences, Paul O'Gorman Leukaemia Research Centre, Gartnavel General Hospital, University of Glasgow, Glasgow, G12 0YN, United Kingdom
| | - Alison M Michie
- School of Cancer Sciences, Paul O'Gorman Leukaemia Research Centre, Gartnavel General Hospital, University of Glasgow, Glasgow, G12 0YN, United Kingdom
| | - Bruno Peault
- MRC Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh, EH16 4UU, United Kingdom
| | - Adam G West
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1QH, United Kingdom
| | - Manuel Salmeron-Sanchez
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, James Watt School of Engineering, The Advanced Research Centre, 11 Chapel Lane, University of Glasgow, Glasgow, G11 6EW, United Kingdom.
| | - Matthew J Dalby
- Centre for the Cellular Microenvironment, School of Molecular Biosciences, The Advanced Research Centre, 11 Chapel Lane, University of Glasgow, Glasgow, G11 6EW, United Kingdom.
| |
Collapse
|
18
|
Hofmann J, Kokkaliaris KD. Bone marrow niches for hematopoietic stem cells: life span dynamics and adaptation to acute stress. Blood 2024; 144:21-34. [PMID: 38579285 DOI: 10.1182/blood.2023023788] [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: 01/18/2024] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 04/07/2024] Open
Abstract
ABSTRACT Hematopoietic stem cells (HSCs) are instrumental for organismal survival because they are responsible for lifelong production of mature blood lineages in homeostasis and response to external stress. To fulfill their function, HSCs rely on reciprocal interactions with specialized tissue microenvironments, termed HSC niches. From embryonic development to advanced aging, HSCs transition through several hematopoietic organs in which they are supported by distinct extrinsic cues. Here, we describe recent discoveries on how HSC niches collectively adapt to ensure robust hematopoietic function during biological aging and after exposure to acute stress. We also discuss the latest strategies leveraging niche-derived signals to revert aging-associated phenotypes and enhance hematopoietic recovery after myeloablation.
Collapse
Affiliation(s)
- Johanna Hofmann
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
- Department 15, Biosciences, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Konstantinos D Kokkaliaris
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Quantitative Spatial Cancer Biology Laboratory, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt am Main, Germany
- University Cancer Center, Frankfurt am Main, Germany
| |
Collapse
|
19
|
Wang HJ, Wang Y, Mirjavadi SS, Andersen T, Moldovan L, Vatankhah P, Russell B, Jin J, Zhou Z, Li Q, Cox CD, Su QP, Ju LA. Microscale geometrical modulation of PIEZO1 mediated mechanosensing through cytoskeletal redistribution. Nat Commun 2024; 15:5521. [PMID: 38951553 PMCID: PMC11217425 DOI: 10.1038/s41467-024-49833-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 06/20/2024] [Indexed: 07/03/2024] Open
Abstract
The microgeometry of the cellular microenvironment profoundly impacts cellular behaviors, yet the link between it and the ubiquitously expressed mechanosensitive ion channel PIEZO1 remains unclear. Herein, we describe a fluorescent micropipette aspiration assay that allows for simultaneous visualization of intracellular calcium dynamics and cytoskeletal architecture in real-time, under varied micropipette geometries. By integrating elastic shell finite element analysis with fluorescent lifetime imaging microscopy and employing PIEZO1-specific transgenic red blood cells and HEK cell lines, we demonstrate a direct correlation between the microscale geometry of aspiration and PIEZO1-mediated calcium signaling. We reveal that increased micropipette tip angles and physical constrictions lead to a significant reorganization of F-actin, accumulation at the aspirated cell neck, and subsequently amplify the tension stress at the dome of the cell to induce more PIEZO1's activity. Disruption of the F-actin network or inhibition of its mobility leads to a notable decline in PIEZO1 mediated calcium influx, underscoring its critical role in cellular mechanosensing amidst geometrical constraints.
Collapse
Affiliation(s)
- Haoqing Jerry Wang
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- Heart Research Institute, Camperdown, Newtown, NSW, 2042, Australia
| | - Yao Wang
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Seyed Sajad Mirjavadi
- School of Aerospace, Mechanical and Mechatronic Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Tomas Andersen
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Laura Moldovan
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- Heart Research Institute, Camperdown, Newtown, NSW, 2042, Australia
| | - Parham Vatankhah
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Blake Russell
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Jasmine Jin
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Zijing Zhou
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, NSW, 2010, Australia
| | - Qing Li
- School of Aerospace, Mechanical and Mechatronic Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Charles D Cox
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Sydney, NSW, 2010, Australia
- Faculty of Medicine, St. Vincent's Clinical School, University of New South Wale, Sydney, NSW, 2010, Australia
| | - Qian Peter Su
- Heart Research Institute, Camperdown, Newtown, NSW, 2042, Australia.
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia.
| | - Lining Arnold Ju
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia.
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia.
- Heart Research Institute, Camperdown, Newtown, NSW, 2042, Australia.
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, 2006, Australia.
| |
Collapse
|
20
|
Lei L, Wen Z, Cao M, Zhang H, Ling SKK, Fu BSC, Qin L, Xu J, Yung PSH. The emerging role of Piezo1 in the musculoskeletal system and disease. Theranostics 2024; 14:3963-3983. [PMID: 38994033 PMCID: PMC11234281 DOI: 10.7150/thno.96959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 05/15/2024] [Indexed: 07/13/2024] Open
Abstract
Piezo1, a mechanosensitive ion channel, has emerged as a key player in translating mechanical stimuli into biological signaling. Its involvement extends beyond physiological and pathological processes such as lymphatic vessel development, axon growth, vascular development, immunoregulation, and blood pressure regulation. The musculoskeletal system, responsible for structural support, movement, and homeostasis, has recently attracted attention regarding the significance of Piezo1. This review aims to provide a comprehensive summary of the current research on Piezo1 in the musculoskeletal system, highlighting its impact on bone formation, myogenesis, chondrogenesis, intervertebral disc homeostasis, tendon matrix cross-linking, and physical activity. Additionally, we explore the potential of targeting Piezo1 as a therapeutic approach for musculoskeletal disorders, including osteoporosis, muscle atrophy, intervertebral disc degeneration, and osteoarthritis.
Collapse
Affiliation(s)
- Lei Lei
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhenkang Wen
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Mingde Cao
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Haozhi Zhang
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Samuel Ka-Kin Ling
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Bruma Sai-Chuen Fu
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ling Qin
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- The Sir Yue-Kong Pao Cancer Centre, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
- Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- The Sir Yue-Kong Pao Cancer Centre, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
- Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Patrick Shu-Hang Yung
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
21
|
Stone AP, Rand E, Thornes G, Kay AG, Barnes AL, Hitchcock IS, Genever PG. Extracellular matrices of stromal cell subtypes regulate phenotype and contribute to the stromal microenvironment in vivo. Stem Cell Res Ther 2024; 15:178. [PMID: 38886845 PMCID: PMC11184721 DOI: 10.1186/s13287-024-03786-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 06/09/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Bone marrow stromal cells (BMSCs) are highly heterogeneous, which may reflect their diverse biological functions, including tissue maintenance, haematopoietic support and immune control. The current understanding of the mechanisms that drive the onset and resolution of heterogeneity, and how BMSCs influence other cells in their environment is limited. Here, we determined how the secretome and importantly the extracellular matrix of BMSCs can influence cellular phenotype. METHODS We used two immortalised clonal BMSC lines isolated from the same heterogeneous culture as model stromal subtypes with distinct phenotypic traits; a multipotent stem-cell-like stromal line (Y201) and a nullipotent non-stem cell stromal line (Y202), isolated from the same donor BMSC pool. Label-free quantitative phase imaging was used to track cell morphology and migration of the BMSC lines over 96 h in colony-forming assays. We quantified the secreted factors of each cell line by mass spectrometry and confirmed presence of proteins in human bone marrow by immunofluorescence. RESULTS Transfer of secreted signals from a stem cell to a non-stem cell resulted in a change in morphology and enhanced migration to more closely match stem cell-like features. Mass spectrometry analysis revealed a significant enrichment of extracellular matrix (ECM) proteins in the Y201 stem cell secretome compared to Y202 stromal cells. We confirmed that Y201 produced a more robust ECM in culture compared to Y202. Growth of Y202 on ECM produced by Y201 or Y202 restored migration and fibroblastic morphology, suggesting that it is the deficiency of ECM production that contributes to its phenotype. The proteins periostin and aggrecan, were detected at 71- and 104-fold higher levels in the Y201 versus Y202 secretome and were subsequently identified by immunofluorescence at rare sites on the endosteal surfaces of mouse and human bone, underlying CD271-positive stromal cells. These proteins may represent key non-cellular components of the microenvironment for bona-fide stem cells important for cell maintenance and phenotype in vivo. CONCLUSIONS We identified plasticity in BMSC morphology and migratory characteristics that can be modified through secreted proteins, particularly from multipotent stem cells. Overall, we demonstrate the importance of specific ECM proteins in co-ordination of cellular phenotype and highlight how non-cellular components of the BMSC microenvironment may provide insights into cell population heterogeneity and the role of BMSCs in health and disease.
Collapse
Affiliation(s)
- Andrew P Stone
- Department of Biology, York Biomedical Research Institute, University of York, York, UK.
- Brandeis University, Waltham, MA, USA.
| | - Emma Rand
- Department of Biology, York Biomedical Research Institute, University of York, York, UK
| | - Gabriel Thornes
- Department of Biology, York Biomedical Research Institute, University of York, York, UK
| | - Alasdair G Kay
- Department of Biology, York Biomedical Research Institute, University of York, York, UK
| | - Amanda L Barnes
- Department of Biology, York Biomedical Research Institute, University of York, York, UK
| | - Ian S Hitchcock
- Department of Biology, York Biomedical Research Institute, University of York, York, UK
| | - Paul G Genever
- Department of Biology, York Biomedical Research Institute, University of York, York, UK
| |
Collapse
|
22
|
Carpenter RS, Maryanovich M. Systemic and local regulation of hematopoietic homeostasis in health and disease. NATURE CARDIOVASCULAR RESEARCH 2024; 3:651-665. [PMID: 39196230 DOI: 10.1038/s44161-024-00482-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 04/24/2024] [Indexed: 08/29/2024]
Abstract
Hematopoietic stem cells (HSCs) generate all blood cell lineages responsible for tissue oxygenation, life-long hematopoietic homeostasis and immune protection. In adulthood, HSCs primarily reside in the bone marrow (BM) microenvironment, consisting of diverse cell types that constitute the stem cell 'niche'. The adaptability of the hematopoietic system is required to respond to the needs of the host, whether to maintain normal physiology or during periods of physical, psychosocial or environmental stress. Hematopoietic homeostasis is achieved by intricate coordination of systemic and local factors that orchestrate the function of HSCs throughout life. However, homeostasis is not a static process; it modulates HSC and progenitor activity in response to circadian rhythms coordinated by the central and peripheral nervous systems, inflammatory cues, metabolites and pathologic conditions. Here, we review local and systemic factors that impact hematopoiesis, focusing on the implications of aging, stress and cardiovascular disease.
Collapse
Affiliation(s)
- Randall S Carpenter
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Ruth L. and David S. Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Maria Maryanovich
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA.
- Ruth L. and David S. Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.
- Cancer Dormancy and Tumor Microenvironment Institute, Albert Einstein College of Medicine, Bronx, NY, USA.
- Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
23
|
Cuahtecontzi Delint R, Jaffery H, Ishak MI, Nobbs AH, Su B, Dalby MJ. Mechanotransducive surfaces for enhanced cell osteogenesis, a review. BIOMATERIALS ADVANCES 2024; 160:213861. [PMID: 38663159 DOI: 10.1016/j.bioadv.2024.213861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/31/2024] [Accepted: 04/12/2024] [Indexed: 05/04/2024]
Abstract
Novel strategies employing mechano-transducing materials eliciting biological outcomes have recently emerged for controlling cellular behaviour. Targeted cellular responses are achieved by manipulating physical, chemical, or biochemical modification of material properties. Advances in techniques such as nanopatterning, chemical modification, biochemical molecule embedding, force-tuneable materials, and artificial extracellular matrices are helping understand cellular mechanotransduction. Collectively, these strategies manipulate cellular sensing and regulate signalling cascades including focal adhesions, YAP-TAZ transcription factors, and multiple osteogenic pathways. In this minireview, we are providing a summary of the influence that these materials, particularly titanium-based orthopaedic materials, have on cells. We also highlight recent complementary methodological developments including, but not limited to, the use of metabolomics for identification of active biomolecules that drive cellular differentiation.
Collapse
Affiliation(s)
- Rosalia Cuahtecontzi Delint
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK.
| | - Hussain Jaffery
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Mohd I Ishak
- Bristol Dental School, University of Bristol, Lower Maudlin Street, Bristol BS1 2LY, UK
| | - Angela H Nobbs
- Bristol Dental School, University of Bristol, Lower Maudlin Street, Bristol BS1 2LY, UK
| | - Bo Su
- Bristol Dental School, University of Bristol, Lower Maudlin Street, Bristol BS1 2LY, UK
| | - Matthew J Dalby
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| |
Collapse
|
24
|
Wang F, Wang H, Zhang H, Sun B, Wang Z. A Novel Mechanism of MSCs Responding to Occlusal Force for Bone Homeostasis. J Dent Res 2024; 103:642-651. [PMID: 38665065 DOI: 10.1177/00220345241236120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2024] Open
Abstract
Alveolar bone, as tooth-supporting bone for mastication, is sensitive to occlusal force. However, the mechanism of alveolar bone loss after losing occlusal force remains unclear. Here, we performed single-cell RNA sequencing of nonhematopoietic (CD45-) cells in mouse alveolar bone after removing the occlusal force. Mesenchymal stromal cells (MSCs) and endothelial cell (EC) subsets were significantly decreased in frequency, as confirmed by immunofluorescence and flow cytometry. The osteogenic and proangiogenic abilities of MSCs were impaired, and the expression of mechanotransducers yes associated protein 1 (Yap) and WW domain containing transcription regulator 1 (Taz) in MSCs decreased. Conditional deletion of Yap and Taz from LepR+ cells, which are enriched in MSCs that are important for adult bone homeostasis, significantly decreased alveolar bone mass and resisted any further changes in bone mass induced by occlusal force changes. Interestingly, LepR-Cre; Yapf/f; Tazf/f mice showed a decrease in CD31hi endomucin (Emcn)hi endothelium, and the expression of some EC-derived signals acting on osteoblastic cells was inhibited in alveolar bone. Mechanistically, conditional deletion of Yap and Taz in LepR+ cells inhibited the secretion of pleiotrophin (Ptn), which impaired the proangiogenic capacity of LepR+ cells. Knockdown in MSC-derived Ptn repressed human umbilical vein EC tube formation in vitro. More important, administration of recombinant PTN locally recovered the frequency of CD31hiEmcnhi endothelium and rescued the low bone mass phenotype of LepR-Cre; Yapf/f; Tazf/f mice. Taken together, these findings suggest that occlusal force governs MSC-regulated endothelium to maintain alveolar bone homeostasis through the Yap/Taz/Ptn axis, providing a reference for further understanding of the relationship between dysfunction and bone homeostasis.
Collapse
Affiliation(s)
- F Wang
- Department of Oral Implantology & Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - H Wang
- Department of Oral Implantology & Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - H Zhang
- Department of Oral Implantology & Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - B Sun
- Department of Oral Implantology & Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Z Wang
- Department of Oral Implantology & Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| |
Collapse
|
25
|
Chen Y, Weng Y, Huang J, Li Q, Sun B, Wang H, Wang Z. Leptin receptor (+) stromal cells respond to periodontitis and attenuate alveolar bone repair via CCRL2-mediated Wnt inhibition. J Bone Miner Res 2024; 39:611-626. [PMID: 38477792 DOI: 10.1093/jbmr/zjae036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/14/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024]
Abstract
The impaired bone healing in tooth extraction sockets due to periodontitis presents a major obstacle to restoring oral health. The mechanisms regulating the osteogenic capacity of jawbone-derived stromal cells in the periodontitis microenvironment remain elusive. Leptin receptor (LepR) expressing stromal cells, which largely overlap with Cxcl12-abundant reticular (CAR) cells in bone tissue, rapidly proliferate and differentiate into bone-forming cells during extraction socket healing to support alveolar bone repair. In this study, we identify that CCRL2 is significantly expressed and inhibits osteogenesis in LepR+/CAR cells of alveolar bones with periodontitis. The Ccrl2-KO mice exhibit significant improvements in bone healing in extraction sockets with periodontitis. Specifically, the binding of CCRL2 to SFRP1 on the surface of LepR+/CAR cells can amplify the suppressive effect of SFRP1 on Wnt signaling under inflammation, thus hindering the osteogenic differentiation of LepR+/CAR cells and resulting in poor bone healing in extraction sockets with periodontitis. Together, we clarify that the CCRL2 receptor of LepR+/CAR cells can respond to periodontitis and crosstalk with Wnt signaling to deteriorate extraction socket healing.
Collapse
Affiliation(s)
- Yongliang Chen
- Department of Oral Implantology and Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, China
| | - Yuteng Weng
- Department of Oral Implantology and Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, China
| | - Jie Huang
- Department of Oral Implantology and Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, China
| | - Qin Li
- Department of Oral Implantology and Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, China
| | - Bin Sun
- Department of Oral Implantology and Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, China
| | - Haicheng Wang
- Department of Oral Implantology and Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, China
| | - Zuolin Wang
- Department of Oral Implantology and Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, China
| |
Collapse
|
26
|
Prasad P, Cancelas JA. From Marrow to Bone and Fat: Exploring the Multifaceted Roles of Leptin Receptor Positive Bone Marrow Mesenchymal Stromal Cells. Cells 2024; 13:910. [PMID: 38891042 PMCID: PMC11171870 DOI: 10.3390/cells13110910] [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: 05/05/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/20/2024] Open
Abstract
The bone marrow (BM) stromal cell microenvironment contains non-hematopoietic stromal cells called mesenchymal stromal cells (MSCs). MSCs are plastic adherent, form CFU-Fs, and give rise to osteogenic, adipogenic, chondrogenic progenitors, and most importantly provide HSC niche factor chemokine C-X-C motif ligand 12 (CXCL12) and stem cell factor (SCF). Different authors have defined different markers for mouse MSC identification like PDGFR+Sca-1+ subsets, Nestin+, or LepR+ cells. Of these, the LepR+ cells are the major source of SCF and CXCL12 in the BM microenvironment and play a major role in HSC maintenance and hematopoiesis. LepR+ cells give rise to most of the bones and BM adipocytes, further regulating the microenvironment. In adult BM, LepR+ cells are quiescent but after fracture or irradiation, they proliferate and differentiate into mesenchymal lineage osteogenic, adipogenic and/or chondrogenic cells. They also play a crucial role in the steady-state hematopoiesis process, as well as hematopoietic regeneration and the homing of hematopoietic stem cells (HSCs) after myeloablative injury and/or HSC transplantation. They line the sinusoidal cavities, maintain the trabeculae formation, and provide the space for HSC homing and retention. However, the LepR+ cell subset is heterogeneous; some subsets have higher adipogenic potential, while others express osteollineage-biased genes. Different transcription factors like Early B cell factor 3 (EBF3) or RunX2 help maintain this balance between the self-renewing and committed states, whether osteogenic or adipogenic. The study of LepR+ MSCs holds immense promise for advancing our understanding of HSC biology, tissue regeneration, metabolic disorders, and immune responses. In this review, we will discuss the origin of the BM resident LepR+ cells, different subtypes, and the role of LepR+ cells in maintaining hematopoiesis, osteogenesis, and BM adipogenesis following their multifaceted impact.
Collapse
Affiliation(s)
| | - Jose A. Cancelas
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA;
| |
Collapse
|
27
|
Lin W, Li Q, Liu L, Wang Q, Zhang D, Wang F, Xu R, Fan Y, Xing M, Zhou C, Yuan Q. Early infiltrating NKT lymphocytes attenuate bone regeneration through secretion of CXCL2. SCIENCE ADVANCES 2024; 10:eadl6343. [PMID: 38758783 PMCID: PMC11100573 DOI: 10.1126/sciadv.adl6343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 04/15/2024] [Indexed: 05/19/2024]
Abstract
Trauma rapidly mobilizes the immune response of surrounding tissues and activates regeneration program. Manipulating immune response to promote tissue regeneration shows a broad application prospect. However, the understanding of bone healing dynamics at cellular level remains limited. Here, we characterize the landscape of immune cells after alveolar bone injury and reveal a pivotal role of infiltrating natural killer T (NKT) cells. We observe a rapid increase in NKT cells after injury, which inhibit osteogenic differentiation of mesenchymal stem cells (MSCs) and impair alveolar bone healing. Cxcl2 is up-regulated in NKT cells after injury. Systemic administration of CXCL2-neutralizing antibody or genetic deletion of Cxcl2 improves the bone healing process. In addition, we fabricate a gelatin-based porous hydrogel to deliver NK1.1 depletion antibody, which successfully promotes alveolar bone healing. In summary, our study highlights the importance of NKT cells in the early stage of bone healing and provides a potential therapeutic strategy for accelerating bone regeneration.
Collapse
Affiliation(s)
- Weimin Lin
- State Key Laboratory of Oral Diseases and National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Qiwen Li
- State Key Laboratory of Oral Diseases and National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Linfeng Liu
- State Key Laboratory of Oral Diseases and National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Qian Wang
- State Key Laboratory of Oral Diseases and National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Danting Zhang
- State Key Laboratory of Oral Diseases and National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Feiyu Wang
- State Key Laboratory of Oral Diseases and National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Ruoshi Xu
- State Key Laboratory of Oral Diseases and National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Yi Fan
- State Key Laboratory of Oral Diseases and National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, Winnipeg R3T 2N2, Canada
| | - Chenchen Zhou
- State Key Laboratory of Oral Diseases and National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases and National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, 610041 Chengdu, Sichuan, China
| |
Collapse
|
28
|
Liu YL, Tang XT, Shu HS, Zou W, Zhou BO. Fibrous periosteum repairs bone fracture and maintains the healed bone throughout mouse adulthood. Dev Cell 2024; 59:1192-1209.e6. [PMID: 38554700 DOI: 10.1016/j.devcel.2024.03.019] [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: 04/02/2023] [Revised: 12/07/2023] [Accepted: 03/06/2024] [Indexed: 04/02/2024]
Abstract
Bone is regarded as one of few tissues that heals without fibrous scar. The outer layer of the periosteum is covered with fibrous tissue, whose function in bone formation is unknown. We herein developed a system to distinguish the fate of fibrous-layer periosteal cells (FL-PCs) from the skeletal stem/progenitor cells (SSPCs) in the cambium-layer periosteum and bone marrow in mice. We showed that FL-PCs did not participate in steady-state osteogenesis, but formed the main body of fibrocartilaginous callus during fracture healing. Moreover, FL-PCs invaded the cambium-layer periosteum and bone marrow after fracture, forming neo-SSPCs that continued to maintain the healed bones throughout adulthood. The FL-PC-derived neo-SSPCs expressed lower levels of osteogenic signature genes and displayed lower osteogenic differentiation activity than the preexisting SSPCs. Consistent with this, healed bones were thinner and formed more slowly than normal bones. Thus, the fibrous periosteum becomes the cellular origin of bones after fracture and alters bone properties permanently.
Collapse
Affiliation(s)
- Yiming Liam Liu
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Xinyu Thomas Tang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Hui Sophie Shu
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Weiguo Zou
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; Key Laboratory of RNA Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.
| | - Bo O Zhou
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences, Tianjin 300020, China.
| |
Collapse
|
29
|
Winter S, Götze KS, Hecker JS, Metzeler KH, Guezguez B, Woods K, Medyouf H, Schäffer A, Schmitz M, Wehner R, Glauche I, Roeder I, Rauner M, Hofbauer LC, Platzbecker U. Clonal hematopoiesis and its impact on the aging osteo-hematopoietic niche. Leukemia 2024; 38:936-946. [PMID: 38514772 PMCID: PMC11073997 DOI: 10.1038/s41375-024-02226-6] [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: 11/16/2023] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 03/23/2024]
Abstract
Clonal hematopoiesis (CH) defines a premalignant state predominantly found in older persons that increases the risk of developing hematologic malignancies and age-related inflammatory diseases. However, the risk for malignant transformation or non-malignant disorders is variable and difficult to predict, and defining the clinical relevance of specific candidate driver mutations in individual carriers has proved to be challenging. In addition to the cell-intrinsic mechanisms, mutant cells rely on and alter cell-extrinsic factors from the bone marrow (BM) niche, which complicates the prediction of a mutant cell's fate in a shifting pre-malignant microenvironment. Therefore, identifying the insidious and potentially broad impact of driver mutations on supportive niches and immune function in CH aims to understand the subtle differences that enable driver mutations to yield different clinical outcomes. Here, we review the changes in the aging BM niche and the emerging evidence supporting the concept that CH can progressively alter components of the local BM microenvironment. These alterations may have profound implications for the functionality of the osteo-hematopoietic niche and overall bone health, consequently fostering a conducive environment for the continued development and progression of CH. We also provide an overview of the latest technology developments to study the spatiotemporal dependencies in the CH BM niche, ideally in the context of longitudinal studies following CH over time. Finally, we discuss aspects of CH carrier management in clinical practice, based on work from our group and others.
Collapse
Affiliation(s)
- Susann Winter
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Katharina S Götze
- German Cancer Consortium (DKTK), CHOICE Consortium, Partner Sites Dresden/Munich/Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Medicine III, Technical University of Munich (TUM), School of Medicine and Health, Munich, Germany
- German MDS Study Group (D-MDS), Leipzig, Germany
| | - Judith S Hecker
- German Cancer Consortium (DKTK), CHOICE Consortium, Partner Sites Dresden/Munich/Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Medicine III, Technical University of Munich (TUM), School of Medicine and Health, Munich, Germany
- TranslaTUM, Center for Translational Cancer Research, Technical University of Munich (TUM), Munich, Germany
| | - Klaus H Metzeler
- German Cancer Consortium (DKTK), CHOICE Consortium, Partner Sites Dresden/Munich/Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Disease, University of Leipzig Medical Center, Leipzig, Germany
| | - Borhane Guezguez
- German Cancer Consortium (DKTK), CHOICE Consortium, Partner Sites Dresden/Munich/Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Hematology and Oncology, University Medical Center Mainz, Mainz, Germany
| | - Kevin Woods
- German Cancer Consortium (DKTK), CHOICE Consortium, Partner Sites Dresden/Munich/Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Hematology and Oncology, University Medical Center Mainz, Mainz, Germany
| | - Hind Medyouf
- German Cancer Consortium (DKTK), CHOICE Consortium, Partner Sites Dresden/Munich/Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Frankfurt am Main, Germany
| | - Alexander Schäffer
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
| | - Marc Schmitz
- German Cancer Consortium (DKTK), CHOICE Consortium, Partner Sites Dresden/Munich/Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Rebekka Wehner
- German Cancer Consortium (DKTK), CHOICE Consortium, Partner Sites Dresden/Munich/Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Ingmar Glauche
- Institute for Medical Informatics and Biometry, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Ingo Roeder
- German Cancer Consortium (DKTK), CHOICE Consortium, Partner Sites Dresden/Munich/Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Institute for Medical Informatics and Biometry, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Martina Rauner
- Division of Endocrinology, Diabetes and Bone Diseases, Department of Medicine III, and Center for Healthy Aging, University Medical Center, TU Dresden, Dresden, Germany
| | - Lorenz C Hofbauer
- German Cancer Consortium (DKTK), CHOICE Consortium, Partner Sites Dresden/Munich/Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Division of Endocrinology, Diabetes and Bone Diseases, Department of Medicine III, and Center for Healthy Aging, University Medical Center, TU Dresden, Dresden, Germany.
| | - Uwe Platzbecker
- German Cancer Consortium (DKTK), CHOICE Consortium, Partner Sites Dresden/Munich/Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
- German MDS Study Group (D-MDS), Leipzig, Germany.
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Disease, University of Leipzig Medical Center, Leipzig, Germany.
| |
Collapse
|
30
|
Nie X, Abbasi Y, Chung MK. Piezo1 and Piezo2 collectively regulate jawbone development. Development 2024; 151:dev202386. [PMID: 38619396 PMCID: PMC11128276 DOI: 10.1242/dev.202386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 04/08/2024] [Indexed: 04/16/2024]
Abstract
Piezo1 and Piezo2 are recently reported mechanosensory ion channels that transduce mechanical stimuli from the environment into intracellular biochemical signals in various tissues and organ systems. Here, we show that Piezo1 and Piezo2 display a robust expression during jawbone development. Deletion of Piezo1 in neural crest cells causes jawbone malformations in a small but significant number of mice. We further demonstrate that disruption of Piezo1 and Piezo2 in neural crest cells causes more striking defects in jawbone development than any single knockout, suggesting essential but partially redundant roles of Piezo1 and Piezo2. In addition, we observe defects in other neural crest derivatives such as malformation of the vascular smooth muscle in double knockout mice. Moreover, TUNEL examinations reveal excessive cell death in osteogenic cells of the maxillary and mandibular arches of the double knockout mice, suggesting that Piezo1 and Piezo2 together regulate cell survival during jawbone development. We further demonstrate that Yoda1, a Piezo1 agonist, promotes mineralization in the mandibular arches. Altogether, these data firmly establish that Piezo channels play important roles in regulating jawbone formation and maintenance.
Collapse
Affiliation(s)
- Xuguang Nie
- Department of Neural and Pain Sciences, School of Dentistry, the University of Maryland, Baltimore, MD 21201,USA
| | - Yasaman Abbasi
- Department of Neural and Pain Sciences, School of Dentistry, the University of Maryland, Baltimore, MD 21201,USA
| | - Man-Kyo Chung
- Department of Neural and Pain Sciences, School of Dentistry, the University of Maryland, Baltimore, MD 21201,USA
- Center to Advance Chronic Pain Research, the University of Maryland, Baltimore, MD 21201,USA
| |
Collapse
|
31
|
Haga CL, Booker CN, Strivelli J, Boregowda SV, Phinney DG. Comparative transcriptome analysis of bone marrow resident versus culture-expanded mouse mesenchymal stem/stromal cells. Cytotherapy 2024; 26:498-505. [PMID: 38372680 PMCID: PMC11065607 DOI: 10.1016/j.jcyt.2024.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/26/2024] [Accepted: 01/27/2024] [Indexed: 02/20/2024]
Abstract
BACKGROUND AIMS Mesenchymal stem/stromal cells (MSCs) are defined as culture-expanded populations, and although these cells recapitulate many properties of bone marrow (BM) resident skeletal stem/progenitor cells, few studies have directly compared these populations to evaluate how culture adaptation and expansion impact critical quality attributes. METHODS We analyzed by RNA sequencing Lin-SCA1+ MSCs enriched from BM by immunodepletion (ID) and after subsequent culture expansion (Ex) and Lin-LEPR+ MSCs sorted (S) directly from BM. Pairwise comparisons were used to identify differentially expressed genes (DEGs) between populations, and gene set enrichment analysis was employed to identify biological pathways/processes unique to each population. K-means cluster analysis resolved isolation status-dependent changes in transcription in pseudotime. RESULTS Hierarchical clustering segregated populations by isolation process, and principal component analysis identified transcripts related to vasculature development, ossification and inflammatory/cytokine signaling as key drivers of population variance. Pairwise comparisons identified 3849 DEGs in ID versus S BM-MSCs mapping to Gene Ontology (GO) terms related to immune and metabolic processes and 334 DEGs in Ex versus ID BM-MSCs mapping to GO terms related to tissue development, cell growth and replication and organelle organization. K-means cluster analysis revealed significant differences in transcripts encoding stemness and differentiation markers, extracellular matrix structural constituents and remodeling enzymes and paracrine-acting factors between populations. CONCLUSIONS These comparative analyses reveal significant differences in gene expression signatures between BM resident and culture-expanded MSCs, thereby providing new insight into how culture adaptation/expansion endows the latter with unique quality attributes.
Collapse
Affiliation(s)
- Christopher L Haga
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute of Biomedical Innovation and Technology, Jupiter, Florida, USA
| | - Cori N Booker
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute of Biomedical Innovation and Technology, Jupiter, Florida, USA
| | - Jacqueline Strivelli
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute of Biomedical Innovation and Technology, Jupiter, Florida, USA
| | - Siddaraju V Boregowda
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute of Biomedical Innovation and Technology, Jupiter, Florida, USA
| | - Donald G Phinney
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute of Biomedical Innovation and Technology, Jupiter, Florida, USA.
| |
Collapse
|
32
|
Dai K, Wang J, Liu C. Biomaterial-assisted therapeutic cell production and modification in vivo. Exp Hematol 2024; 133:104192. [PMID: 38432427 DOI: 10.1016/j.exphem.2024.104192] [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: 09/19/2023] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/05/2024]
Abstract
Hematopoietic stem cell transplantation remains the preferred treatment for a variety of hematopoietic function disorders. To address the issue of limited numbers of hematopoietic stem/progenitor cells (HSPCs), significant progress has been made in the technology for ex vivo expansion of HSPCs. In addition, biomaterial-assisted in vivo production technology for therapeutic cells, including HSPCs, is gradually gaining attention. With the aid of specifically functional biomaterials, researchers can construct bone-like tissues exhibiting typical bone marrow-like structures (termed in vivo osteo-organoids in this article) for the production of therapeutic cells. These in vivo osteo-organoids mimic the native bone marrow niche and provide a microenvironment conducive to the expansion and differentiation of HSPCs. In this perspective article, we systematically summarize the history of in vivo osteo-organoids as a model for studying hematopoiesis and cancer metastasis and propose the challenges faced by the in vivo osteo-organoid production platform for therapeutic cells in terms of clinical translation. Ultimately, we hope to achieve functional customization of in vivo osteo-organoid-derived cells through continuously developed material design methods, so as to meet the treatment needs of different types of diseases and bring hope for life to more people.
Collapse
Affiliation(s)
- Kai Dai
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, People's Republic of China; State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, People's Republic of China; Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, People's Republic of China
| | - Jing Wang
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, People's Republic of China; State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, People's Republic of China; Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, People's Republic of China; Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, People's Republic of China.
| | - Changsheng Liu
- Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, People's Republic of China; Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, People's Republic of China; Key Laboratory for Ultrafine Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, People's Republic of China.
| |
Collapse
|
33
|
Chen Q, Wu Z, Shi Y, Li Z, Yang J, Qu M, Zhang S, Wang Z, Ji N, Li J, Shen Y, Xie L, Chen Q. Loss of PA28γ exacerbates imbalanced differentiation of bone marrow stromal cells during bone formation and bone healing in mice. J Bone Miner Res 2024; 39:326-340. [PMID: 38477820 DOI: 10.1093/jbmr/zjae012] [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: 08/24/2023] [Revised: 01/04/2024] [Accepted: 01/10/2024] [Indexed: 03/14/2024]
Abstract
Proteasome activator subunit 3 (PA28γ) is a member of the proteasome activator family, which mainly regulates the degradation and stability of proteins. Studies have shown that it plays crucial roles in lipid formation, stemness maintenance, and blood vessel formation. However, few studies have clarified the association between PA28γ and bone diseases. Herein, we identified PA28γ as a previously unknown regulator of bone homeostasis that coordinates bone formation and lipid accumulation. PA28γ-knockout mice presented with the characteristics of low bone mass and accumulation of lipids. Suppressed expression of PA28γ restrained the osteogenic differentiation and enhanced the adipogenic differentiation of bone marrow stromal cells (BMSCs). Overexpression of PA28γ promoted osteogenic differentiation and inhibited adipogenic differentiation of BMSCs. Mechanistically, PA28γ interacted with Wnt5α, and the two interactors appeared to be positively correlated. PA28γ mainly activated the downstream Wnt/β-catenin signaling pathway, which affects BMSCs differentiation homeostasis. Deletion of Wnt5α significantly delayed the promotion of osteogenic differentiation and partially alleviated the inhibitory effect of adipogenic differentiation of BMSCs in the PA28γ-overexpressing group. Furthermore, we demonstrated that PA28γ-knockout mice had an inhibited rate of bone healing in a drill-hole femoral bone defect model in vivo. Therefore, our results confirm the effects of PA28γ on bone formation and bone defect repair, indicating that PA28γ mainly interacts with Wnt5α to activate the Wnt/β-catenin signaling pathway regulating BMSCs differentiation homeostasis. Our results reveal the function of PA28γ in bone diseases and provide a new theoretical basis for expanding the treatment of bone diseases.
Collapse
Affiliation(s)
- Qian Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, P. R. China
| | - ZuPing Wu
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, P. R. China
| | - YuJie Shi
- Department of Stomatology, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, P. R. China
| | - ZaiYe Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - JiaKang Yang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - MoYuan Qu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - ShiYu Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zheng Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ning Ji
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - YingQiang Shen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Liang Xie
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qianming Chen
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, P. R. China
| |
Collapse
|
34
|
Chen X, Liu C, Wang J, Du C. Hematopoietic Stem Cells as an Integrative Hub Linking Lifestyle to Cardiovascular Health. Cells 2024; 13:712. [PMID: 38667327 PMCID: PMC11049205 DOI: 10.3390/cells13080712] [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: 02/16/2024] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Despite breakthroughs in modern medical care, the incidence of cardiovascular disease (CVD) is even more prevalent globally. Increasing epidemiologic evidence indicates that emerging cardiovascular risk factors arising from the modern lifestyle, including psychosocial stress, sleep problems, unhealthy diet patterns, physical inactivity/sedentary behavior, alcohol consumption, and tobacco smoking, contribute significantly to this worldwide epidemic, while its underpinning mechanisms are enigmatic. Hematological and immune systems were recently demonstrated to play integrative roles in linking lifestyle to cardiovascular health. In particular, alterations in hematopoietic stem cell (HSC) homeostasis, which is usually characterized by proliferation, expansion, mobilization, megakaryocyte/myeloid-biased differentiation, and/or the pro-inflammatory priming of HSCs, have been shown to be involved in the persistent overproduction of pro-inflammatory myeloid leukocytes and platelets, the cellular protagonists of cardiovascular inflammation and thrombosis, respectively. Furthermore, certain lifestyle factors, such as a healthy diet pattern and physical exercise, have been documented to exert cardiovascular protective effects through promoting quiescence, bone marrow retention, balanced differentiation, and/or the anti-inflammatory priming of HSCs. Here, we review the current understanding of and progression in research on the mechanistic interrelationships among lifestyle, HSC homeostasis, and cardiovascular health. Given that adhering to a healthy lifestyle has become a mainstream primary preventative approach to lowering the cardiovascular burden, unmasking the causal links between lifestyle and cardiovascular health from the perspective of hematopoiesis would open new opportunities to prevent and treat CVD in the present age.
Collapse
Affiliation(s)
| | | | - Junping Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China; (X.C.); (C.L.)
| | - Changhong Du
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China; (X.C.); (C.L.)
| |
Collapse
|
35
|
Matteini F, Montserrat-Vazquez S, Florian MC. Rejuvenating aged stem cells: therapeutic strategies to extend health and lifespan. FEBS Lett 2024. [PMID: 38604982 DOI: 10.1002/1873-3468.14865] [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: 10/23/2023] [Revised: 02/03/2024] [Accepted: 03/07/2024] [Indexed: 04/13/2024]
Abstract
Aging is associated with a global decline in stem cell function. To date, several strategies have been proposed to rejuvenate aged stem cells: most of these result in functional improvement of the tissue where the stem cells reside, but the impact on the lifespan of the whole organism has been less clearly established. Here, we review some of the most recent work dealing with interventions that improve the regenerative capacity of aged somatic stem cells in mammals and that might have important translational possibilities. Overall, we underscore that somatic stem cell rejuvenation represents a strategy to improve tissue homeostasis upon aging and present some recent approaches with the potential to affect health span and lifespan of the whole organism.
Collapse
Affiliation(s)
- Francesca Matteini
- Stem Cell Aging Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), Barcelona, Spain
- Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P-CMR[C]), Barcelona, Spain
| | - Sara Montserrat-Vazquez
- Stem Cell Aging Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), Barcelona, Spain
- Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P-CMR[C]), Barcelona, Spain
| | - M Carolina Florian
- Stem Cell Aging Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), Barcelona, Spain
- Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P-CMR[C]), Barcelona, Spain
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
- The Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| |
Collapse
|
36
|
Dong R, Wei J, Tian S, Wang J, Ma Y, Li Y, Liu RX, Liu YQ. Single-cell RNA transcriptomics reveals Du-Zhong-Wan promotes osteoporotic fracture healing via YAP/β-catenin/VEGF axis in BMSCs. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:155572. [PMID: 39366157 DOI: 10.1016/j.phymed.2024.155572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/29/2024] [Accepted: 03/27/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND Our previous study demonstrated that Du-Zhong-Wan (DZW) promoted osteoporotic fracture (OPF) healing by enhancing osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and angiogenesis of endothelial cells (ECs). However, the heterogeneity of BMSCs and ECs, as well as the specific molecular mechanism underlying these effects, still require further evaluation. PURPOSE The primary objective of this study was to elucidate the heterogeneity of BMSCs and ECs, as well as the cellular-level mechanism of DZW against OPF through single-cell RNA sequencing. METHODS In this study, we presented a single-cell atlas of mouse femoral callus, comparing samples with and without DZW treatment, utilizing single-cell RNA sequencing. Variable genes were identified using the FindVariableGenes (FVG) and principal component analysis (PCA) analysis. Additionally, uniform manifold approximation and projection (U-MAP) was employed to reduce and visualize the distinct subclusters. The CellPhoneDB2 method was employed to analyze intercellular communication and quantify the interaction between ligands and receptors within distinct cell clusters. The osteogenic differentiation capacity of BMSCs was assessed by micro-CT, alkaline phosphatase (ALP), and alizarin red S (ARS) assay. The scratch wound assay and tube formation assay were utilized to assess the angiogenic capabilities of ECs in vitro. Additionally, western blot and immunofluorescence experiments were utilized to elucidate the related protein expression. RESULTS Consistent with our previous studies, DZW obviously promoted osteoporotic fracture healing. Moreover, this study discovered 14 cell clusters at the femoral fracture callus, where the BMSCs most actively interacted with ECs, through single-cell sequencing. Notably, DZW significantly elevated the proportion of Lepr+ BMSCs and Podxl+ ECs subgroup, which were respectively considered essential cells for osteoblastogenesis and angiogenesis of arteriolar vessels. The increased proportion of Podxl+ ECs was partially attributed to vascular endothelial growth factor (VEGF), secreted by BMSCs, which were able to be reversed by YAP pharmacological inhibitor verteporfin. Furthermore, the western blot assay revealed elevated expression levels of YAP/β-catenin, VEGF, RUNX2, and OCN in BMSCs treated with DZW, which were counteracted by verteporfin. CONCLUSION The data above indicates that DZW elevates the proportion of LEPR+ BMSCs and Podxl+ ECs, therefore contributing for the osteogenic ability of BMSCs and BMSCs-mediated angiogenesis via activation of the YAP/β-catenin/VEGF axis, which provides novel potential targets and mechanism for DZW in treating OPF in sub-clusters and molecular level.
Collapse
Affiliation(s)
- Renchao Dong
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jun Wei
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shuo Tian
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jie Wang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yu Ma
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yilin Li
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Rui-Xia Liu
- The First Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Yan-Qiu Liu
- Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
37
|
Xu C, Xie X, Shi P, Xue K, Li Y, Wu Y, Wang J. LepR-expressing cells are a critical population in periodontal healing post periodontitis. J Bone Miner Res 2024; 39:59-72. [PMID: 38630879 DOI: 10.1093/jbmr/zjad008] [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: 07/28/2023] [Revised: 11/12/2023] [Accepted: 11/17/2023] [Indexed: 04/19/2024]
Abstract
Identification of promising seed cells plays a pivotal role in achieving tissue regeneration. This study demonstrated that LepR-expressing cells (LepR+ cells) are required for maintaining periodontal homeostasis at the adult stage. We further investigated how LepR+ cells behave in periodontal healing using a ligature-induced periodontitis (PD) and a self-healing murine model with LepRCre/+; R26RtdTomato/+ mice. Lineage tracing experiments revealed that the largely suppressed osteogenic ability of LepR+ cells results from periodontal inflammation. Periodontal defects were partially recovered when the ligature was removed, in which the osteogenic differentiation of LepR+ cell lineage was promoted and contributed to the newly formed alveolar bone. A cell ablation model established with LepRCre/+; R26RtdTomato/+; R26RDTA/+ mice further proved that LepR+ cells are an important cell source of newly formed alveolar bone. Expressions of β-catenin and LEF1 in LepR+ cells were upregulated when the inflammatory stimuli were removed, which are consistent with the functional changes observed during periodontal healing. Furthermore, the conditional upregulation of WNT signaling or the application of sclerostin neutralized antibody promoted the osteogenic function of LepR+ cells. In contrast, the specific knockdown of β-catenin in LepR+ human periodontal ligament cells with small interfering RNA caused arrested osteogenic function. Our findings identified the LepR+ cell lineage as a critical cell population for endogenous periodontal healing post PD, which is regulated by the WNT signaling pathway, making it a promising seed cell population in periodontal tissue regeneration.
Collapse
Affiliation(s)
- Chunmei Xu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xudong Xie
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Peilei Shi
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Kun Xue
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yue Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yafei Wu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Jun Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
38
|
Wu Q, Zhang J, Kumar S, Shen S, Kincaid M, Johnson CB, Zhang YS, Turcotte R, Alt C, Ito K, Homan S, Sherman BE, Shao TY, Slaughter A, Weinhaus B, Song B, Filippi MD, Grimes HL, Lin CP, Ito K, Way SS, Kofron JM, Lucas D. Resilient anatomy and local plasticity of naive and stress haematopoiesis. Nature 2024; 627:839-846. [PMID: 38509363 PMCID: PMC10972750 DOI: 10.1038/s41586-024-07186-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 02/09/2024] [Indexed: 03/22/2024]
Abstract
The bone marrow adjusts blood cell production to meet physiological demands in response to insults. The spatial organization of normal and stress responses are unknown owing to the lack of methods to visualize most steps of blood production. Here we develop strategies to image multipotent haematopoiesis, erythropoiesis and lymphopoiesis in mice. We combine these with imaging of myelopoiesis1 to define the anatomy of normal and stress haematopoiesis. In the steady state, across the skeleton, single stem cells and multipotent progenitors distribute through the marrow enriched near megakaryocytes. Lineage-committed progenitors are recruited to blood vessels, where they contribute to lineage-specific microanatomical structures composed of progenitors and immature cells, which function as the production sites for each major blood lineage. This overall anatomy is resilient to insults, as it was maintained after haemorrhage, systemic bacterial infection and granulocyte colony-stimulating factor (G-CSF) treatment, and during ageing. Production sites enable haematopoietic plasticity as they differentially and selectively modulate their numbers and output in response to insults. We found that stress responses are variable across the skeleton: the tibia and the sternum respond in opposite ways to G-CSF, and the skull does not increase erythropoiesis after haemorrhage. Our studies enable in situ analyses of haematopoiesis, define the anatomy of normal and stress responses, identify discrete microanatomical production sites that confer plasticity to haematopoiesis, and uncover unprecedented heterogeneity of stress responses across the skeleton.
Collapse
Affiliation(s)
- Qingqing Wu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Jizhou Zhang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Sumit Kumar
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Siyu Shen
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Morgan Kincaid
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Courtney B Johnson
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Yanan Sophia Zhang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Raphaël Turcotte
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ruth L. and David S. Gottesman Institute for Stem Cell, Regenerative Medicine Research, Department of Cell Biology and Stem Cell Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Clemens Alt
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Kyoko Ito
- Ruth L. and David S. Gottesman Institute for Stem Cell, Regenerative Medicine Research, Department of Cell Biology and Stem Cell Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Shelli Homan
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Bryan E Sherman
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Tzu-Yu Shao
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Anastasiya Slaughter
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Benjamin Weinhaus
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Baobao Song
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Immunobiology and Center for Systems Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Marie Dominique Filippi
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - H Leighton Grimes
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Immunobiology and Center for Systems Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Charles P Lin
- Advanced Microscopy Program, Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Keisuke Ito
- Ruth L. and David S. Gottesman Institute for Stem Cell, Regenerative Medicine Research, Department of Cell Biology and Stem Cell Institute, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine, Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sing Sing Way
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - J Matthew Kofron
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Daniel Lucas
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
39
|
Collins JM, Lang A, Parisi C, Moharrer Y, Nijsure MP, Thomas Kim JH, Ahmed S, Szeto GL, Qin L, Gottardi R, Dyment NA, Nowlan NC, Boerckel JD. YAP and TAZ couple osteoblast precursor mobilization to angiogenesis and mechanoregulation in murine bone development. Dev Cell 2024; 59:211-227.e5. [PMID: 38141609 PMCID: PMC10843704 DOI: 10.1016/j.devcel.2023.11.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/07/2023] [Accepted: 11/30/2023] [Indexed: 12/25/2023]
Abstract
Fetal bone development occurs through the conversion of avascular cartilage to vascularized bone at the growth plate. This requires coordinated mobilization of osteoblast precursors with blood vessels. In adult bone, vessel-adjacent osteoblast precursors are maintained by mechanical stimuli; however, the mechanisms by which these cells mobilize and respond to mechanical cues during embryonic development are unknown. Here, we show that the mechanoresponsive transcriptional regulators Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) spatially couple osteoblast precursor mobilization to angiogenesis, regulate vascular morphogenesis to control cartilage remodeling, and mediate mechanoregulation of embryonic murine osteogenesis. Mechanistically, YAP and TAZ regulate a subset of osteoblast-lineage cells, identified by single-cell RNA sequencing as vessel-associated osteoblast precursors, which regulate transcriptional programs that direct blood vessel invasion through collagen-integrin interactions and Cxcl12. Functionally, in 3D human cell co-culture, CXCL12 treatment rescues angiogenesis impaired by stromal cell YAP/TAZ depletion. Together, these data establish functions of the vessel-associated osteoblast precursors in bone development.
Collapse
Affiliation(s)
- Joseph M Collins
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Annemarie Lang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cristian Parisi
- Department of Bioengineering, Imperial College London, London, UK
| | - Yasaman Moharrer
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Department of Mechanical Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Madhura P Nijsure
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jong Hyun Thomas Kim
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Saima Ahmed
- Department of Bioengineering, Imperial College London, London, UK
| | | | - Ling Qin
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Riccardo Gottardi
- Department of Pediatrics, Division of Pulmonary Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nathaniel A Dyment
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Niamh C Nowlan
- Department of Bioengineering, Imperial College London, London, UK; School of Mechanical and Materials Engineering, University College Dublin, Dublin, Ireland; UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Joel D Boerckel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
40
|
Xia K, Chen X, Wang W, Liu Q, Zhao M, Ma J, Jia H. Roles of mechanosensitive ion channels in immune cells. Heliyon 2024; 10:e23318. [PMID: 38148826 PMCID: PMC10750075 DOI: 10.1016/j.heliyon.2023.e23318] [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: 09/16/2023] [Revised: 11/25/2023] [Accepted: 11/30/2023] [Indexed: 12/28/2023] Open
Abstract
Mechanosensitive ion channels are a class of membrane-integrated proteins that convert externalmechanical forces, including stretching, pressure, gravity, and osmotic pressure changes, some of which can be caused by pathogen invasion, into electrical and chemical signals transmitted to the cytoplasm. In recent years, with the identification of many of these channels, their roles in the initiation and progression of many diseases have been gradually revealed. Multiple studies have shown that mechanosensitive ion channels regulate the proliferation, activation, and inflammatory responses of immune cells by being expressed on the surface of immune cells and further responding to mechanical forces. Nonetheless, further clarification is required regarding the signaling pathways of immune-cell pattern-recognition receptors and on the impact of microenvironmental changes and mechanical forces on immune cells. This review summarizes the roles of mechanosensitive ion channels in immune cells.
Collapse
Affiliation(s)
- Kexin Xia
- Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Xiaolin Chen
- Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Wenyan Wang
- Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Qianwen Liu
- Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Mai Zhao
- Department of Orthopedic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Haining Road 100, Shanghai, 200080, China
| | - Jiacheng Ma
- The Department of Information Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, 999077, China
| | - Hao Jia
- Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry & Molecular Cellular Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| |
Collapse
|
41
|
Amado NG, Nosyreva ED, Thompson D, Egeland TJ, Ogujiofor OW, Yang M, Fusco AN, Passoni N, Mathews J, Cantarel B, Baker LA, Syeda R. PIEZO1 loss-of-function compound heterozygous mutations in the rare congenital human disorder Prune Belly Syndrome. Nat Commun 2024; 15:339. [PMID: 38184690 PMCID: PMC10771463 DOI: 10.1038/s41467-023-44594-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 12/21/2023] [Indexed: 01/08/2024] Open
Abstract
Prune belly syndrome (PBS), also known as Eagle-Barret syndrome, is a rare, multi-system congenital myopathy primarily affecting males. Phenotypically, PBS cases manifest three cardinal pathological features: urinary tract dilation with poorly contractile smooth muscle, wrinkled flaccid ventral abdominal wall with skeletal muscle deficiency, and intra-abdominal undescended testes. Genetically, PBS is poorly understood. After performing whole exome sequencing in PBS patients, we identify one compound heterozygous variant in the PIEZO1 gene. PIEZO1 is a cation-selective channel activated by various mechanical forces and widely expressed throughout the lower urinary tract. Here we conduct an extensive functional analysis of the PIEZO1 PBS variants that reveal loss-of-function characteristics in the pressure-induced normalized open probability (NPo) of the channel, while no change is observed in single-channel currents. Furthermore, Yoda1, a PIEZO1 activator, can rescue the NPo defect of the PBS mutant channels. Thus, PIEZO1 mutations may be causal for PBS and the in vitro cellular pathophysiological phenotype could be rescued by the small molecule, Yoda1. Activation of PIEZO1 might provide a promising means of treating PBS and other related bladder dysfunctional states.
Collapse
Affiliation(s)
- Nathalia G Amado
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Elena D Nosyreva
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - David Thompson
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Thomas J Egeland
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Osita W Ogujiofor
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Michelle Yang
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alexandria N Fusco
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Niccolo Passoni
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jeremy Mathews
- Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Brandi Cantarel
- Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Linda A Baker
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- The Kidney and Urinary Tract Center, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.
| | - Ruhma Syeda
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
42
|
Hu H, Luo S, Lai P, Lai M, Mao L, Zhang S, Jiang Y, Wen J, Zhou W, Liu X, Wang L, Huang M, Hu Y, Zhao X, Xia L, Zhou W, Jiang Y, Zou Z, Liu A, Guo B, Bai X. ANGPTL4 binds to the leptin receptor to regulate ectopic bone formation. Proc Natl Acad Sci U S A 2024; 121:e2310685120. [PMID: 38147550 PMCID: PMC10769826 DOI: 10.1073/pnas.2310685120] [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: 06/25/2023] [Accepted: 11/17/2023] [Indexed: 12/28/2023] Open
Abstract
Leptin protein was thought to be unique to leptin receptor (LepR), but the phenotypes of mice with mutation in LepR [db/db (diabetes)] and leptin [ob/ob (obese)] are not identical, and the cause remains unclear. Here, we show that db/db, but not ob/ob, mice had defect in tenotomy-induced heterotopic ossification (HO), implicating alternative ligand(s) for LepR might be involved. Ligand screening revealed that ANGPTL4 (angiopoietin-like protein 4), a stress and fasting-induced factor, was elicited from brown adipose tissue after tenotomy, bound to LepR on PRRX1+ mesenchymal cells at the HO site, thus promotes chondrogenesis and HO development. Disruption of LepR in PRRX1+ cells, or lineage ablation of LepR+ cells, or deletion of ANGPTL4 impeded chondrogenesis and HO in mice. Together, these findings identify ANGPTL4 as a ligand for LepR to regulate the formation of acquired HO.
Collapse
Affiliation(s)
- Hongling Hu
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong510630, China
- Department of Trauma and Joint Surgery, Shunde Hospital, Southern Medical University, Foshan, Guangdong528300, China
| | - Sheng Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Pinglin Lai
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong510630, China
| | - Mingqiang Lai
- Department of Orthopaedics, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong510900, China
| | - Linlin Mao
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Sheng Zhang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Yuanjun Jiang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Jiaxin Wen
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Wu Zhou
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Xiaolin Liu
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Liang Wang
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong510630, China
| | - Minjun Huang
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong510630, China
| | - Yanjun Hu
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Xiaoyang Zhao
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Laixin Xia
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Weijie Zhou
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Yu Jiang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA15261
| | - Zhipeng Zou
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Anling Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong510515, China
| | - Bin Guo
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong510515, China
- Department of Orthopaedics, The Tenth Affiliated Hospital, Southern Medical University, Dongguan, Guangdong523018, China
| | - Xiaochun Bai
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong510630, China
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong510515, China
| |
Collapse
|
43
|
Wang W, Zheng X, Wang H, Zuo B, Chen S, Li J. Mechanical Unloading Promotes Osteoclastic Differentiation and Bone Resorption by Modulating the MSC Secretome to Favor Inflammation. Cell Transplant 2024; 33:9636897241236584. [PMID: 38501500 PMCID: PMC10953070 DOI: 10.1177/09636897241236584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 02/11/2024] [Accepted: 02/13/2024] [Indexed: 03/20/2024] Open
Abstract
Aging, space flight, and prolonged bed rest have all been linked to bone loss, and no effective treatments are clinically available at present. Here, with the rodent hindlimb unloading (HU) model, we report that the bone marrow (BM) microenvironment was significantly altered, with an increased number of myeloid cells and elevated inflammatory cytokines. In such inflammatory BM, the osteoclast-mediated bone resorption was greatly enhanced, leading to a shifted bone remodeling balance that ultimately ends up with disuse-induced osteoporosis. Using Piezo1 conditional knockout (KO) mice (Piezo1fl/fl;LepRCre), we proved that lack of mechanical stimuli on LepR+ mesenchymal stem cells (MSCs) is the main reason for the pathological BM inflammation. Mechanically, the secretome of MSCs was regulated by mechanical stimuli. Inadequate mechanical load leads to increased production of inflammatory cytokines, such as interleukin (IL)-1α, IL-6, macrophage colony-stimulating factor 1 (M-CSF-1), and so on, which promotes monocyte proliferation and osteoclastic differentiation. Interestingly, transplantation of 10% cyclic mechanical stretch (CMS)-treated MSCs into HU animals significantly alleviated the BM microenvironment and rebalanced bone remodeling. In summary, our research revealed a new mechanism underlying mechanical unloading-induced bone loss and suggested a novel stem cell-based therapy to potentially prevent disuse-induced osteoporosis.
Collapse
Affiliation(s)
- Wanyuji Wang
- Department of Cell Biology, Zunyi Medical University, Zunyi, China
| | - Xueling Zheng
- Department of Cell Biology, Zunyi Medical University, Zunyi, China
| | - Hehe Wang
- Department of Cell Biology, Zunyi Medical University, Zunyi, China
| | - Bin Zuo
- Department of Orthopedic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Sisi Chen
- Department of Orthopedic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiao Li
- Department of Cell Biology, Zunyi Medical University, Zunyi, China
| |
Collapse
|
44
|
Zhang Y, Liu F. The evolving views of hematopoiesis: from embryo to adulthood and from in vivo to in vitro. J Genet Genomics 2024; 51:3-15. [PMID: 37734711 DOI: 10.1016/j.jgg.2023.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/13/2023] [Accepted: 09/13/2023] [Indexed: 09/23/2023]
Abstract
The hematopoietic system composed of hematopoietic stem and progenitor cells (HSPCs) and their differentiated lineages serves as an ideal model to uncover generic principles of cell fate transitions. From gastrulation onwards, there successively emerge primitive hematopoiesis (that produces specialized hematopoietic cells), pro-definitive hematopoiesis (that produces lineage-restricted progenitor cells), and definitive hematopoiesis (that produces multipotent HSPCs). These nascent lineages develop in several transient hematopoietic sites and finally colonize into lifelong hematopoietic sites. The development and maintenance of hematopoietic lineages are orchestrated by cell-intrinsic gene regulatory networks and cell-extrinsic microenvironmental cues. Owing to the progressive methodology (e.g., high-throughput lineage tracing and single-cell functional and omics analyses), our understanding of the developmental origin of hematopoietic lineages and functional properties of certain hematopoietic organs has been updated; meanwhile, new paradigms to characterize rare cell types, cell heterogeneity and its causes, and comprehensive regulatory landscapes have been provided. Here, we review the evolving views of HSPC biology during developmental and postnatal hematopoiesis. Moreover, we discuss recent advances in the in vitro induction and expansion of HSPCs, with a focus on the implications for clinical applications.
Collapse
Affiliation(s)
- Yifan Zhang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
| | - Feng Liu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China; State Key Laboratory of Membrane Biology, Institute of Zoology, Institute for Stem Cell and Regeneration, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
45
|
Singh AK, Prasad P, Cancelas JA. Mesenchymal stromal cells, metabolism, and mitochondrial transfer in bone marrow normal and malignant hematopoiesis. Front Cell Dev Biol 2023; 11:1325291. [PMID: 38169927 PMCID: PMC10759248 DOI: 10.3389/fcell.2023.1325291] [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: 10/21/2023] [Accepted: 11/23/2023] [Indexed: 01/05/2024] Open
Abstract
Hematopoietic stem cell (HSC) transplantation-based treatments are in different phases of clinical development, ranging from current therapies to a promise in the repair and regeneration of diseased tissues and organs. Mesenchymal stromal/stem cells (MSCs), which are fibroblast-like heterogeneous progenitors with multilineage differentiation (osteogenic, chondrogenic, and adipogenic) and self-renewal potential, and exist in the bone marrow (BM), adipose, and synovium, among other tissues, represent one of the most widely used sources of stem cells in regenerative medicine. MSCs derived from bone marrow (BM-MSCs) exhibit a variety of traits, including the potential to drive HSC fate and anti-inflammatory and immunosuppressive capabilities via paracrine activities and interactions with the innate and adaptive immune systems. The role of BM-MSC-derived adipocytes is more controversial and may act as positive or negative regulators of benign or malignant hematopoiesis based on their anatomical location and functional crosstalk with surrounding cells in the BM microenvironment. This review highlights the most recent clinical and pre-clinical findings on how BM-MSCs interact with the surrounding HSCs, progenitors, and immune cells, and address some recent insights on the mechanisms that mediate MSCs and adipocyte metabolic control through a metabolic crosstalk between BM microenvironment cells and intercellular mitochondrial transfer in normal and malignant hematopoiesis.
Collapse
Affiliation(s)
- Abhishek K. Singh
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Parash Prasad
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Jose A. Cancelas
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
46
|
Ye YJ, Xu YF, Hou YB, Yin DC, Su DB, Zhao ZX. Regulation of Tendon Stem Cell Behavior by Designed Nanoporous Topography of Microfibers. Biomacromolecules 2023; 24:5859-5870. [PMID: 38015033 DOI: 10.1021/acs.biomac.3c00896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Nano scale topography scaffold is more bioactive and biomimetic than smooth fiber topographies. Tendon stem cells (TSCs) play important roles in the tendinogenesis of tendon tissue engineering, but the effects and mechanisms of nano topography on TSC behavior are still unclear. This study determined whether the morphology, proliferation, cytoskeleton, and differentiation of TSCs are affected by topography of scaffold in vitro. The porous PA56 scaffolds were prepared with different concentration ratios of glycerol as the molecular template by electrospinning. Its topological characteristics, hydrophilicity, and degradation properties varied with glycerol proportion and movement rate of the receiving plate. Porous fibers promoted the proliferation of TSCs and the number of TSCs varied with topography. Although there was no significant difference due to the small sample size, the number of pseudopodia and cell polarizability still showed differences among different topographies. The morphology of actin cytoskeleton of TSCs showed difference among cultured on porous fibers, smooth fibers, and in culture media with no fiber, suggesting the orientation growth of cells on porous fiber. Moreover, porous fibers promoted teno-lineage differentiation of TSCs by upregulating tendon-specific gene expression. These findings provide evidence that nano porous topography scaffold promotes TSC proliferation, cytoskeleton orientation, and tenogenic differentiation.
Collapse
Affiliation(s)
- Ya-Jing Ye
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, P. R. China
| | - Yi-Fan Xu
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, P. R. China
| | - Ya-Bo Hou
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, P. R. China
| | - Da-Chuan Yin
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, P. R. China
| | - Dan-Bo Su
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, P. R. China
| | - Zi-Xu Zhao
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, Shaanxi, P. R. China
| |
Collapse
|
47
|
Liu H, Jiang H, Liu X, Wang X. Physicochemical understanding of biomineralization by molecular vibrational spectroscopy: From mechanism to nature. EXPLORATION (BEIJING, CHINA) 2023; 3:20230033. [PMID: 38264681 PMCID: PMC10742219 DOI: 10.1002/exp.20230033] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 06/25/2023] [Indexed: 01/25/2024]
Abstract
The process and mechanism of biomineralization and relevant physicochemical properties of mineral crystals are remarkably sophisticated multidisciplinary fields that include biology, chemistry, physics, and materials science. The components of the organic matter, structural construction of minerals, and related mechanical interaction, etc., could help to reveal the unique nature of the special mineralization process. Herein, the paper provides an overview of the biomineralization process from the perspective of molecular vibrational spectroscopy, including the physicochemical properties of biomineralized tissues, from physiological to applied mineralization. These physicochemical characteristics closely to the hierarchical mineralization process include biological crystal defects, chemical bonding, atomic doping, structural changes, and content changes in organic matter, along with the interface between biocrystals and organic matter as well as the specific mechanical effects for hardness and toughness. Based on those observations, the special physiological properties of mineralization for enamel and bone, as well as the possible mechanism of pathological mineralization and calcification such as atherosclerosis, tumor micro mineralization, and urolithiasis are also reviewed and discussed. Indeed, the clearly defined physicochemical properties of mineral crystals could pave the way for studies on the mechanisms and applications.
Collapse
Affiliation(s)
- Hao Liu
- State Key Laboratory of Digital Medical EngineeringSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingJiangsuChina
| | - Hui Jiang
- State Key Laboratory of Digital Medical EngineeringSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingJiangsuChina
| | - Xiaohui Liu
- State Key Laboratory of Digital Medical EngineeringSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingJiangsuChina
| | - Xuemei Wang
- State Key Laboratory of Digital Medical EngineeringSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingJiangsuChina
| |
Collapse
|
48
|
Gao X, Murphy MM, Peyer JG, Ni Y, Yang M, Zhang Y, Guo J, Kara N, Embree C, Tasdogan A, Ubellacker JM, Crane GM, Fang S, Zhao Z, Shen B, Morrison SJ. Leptin receptor + cells promote bone marrow innervation and regeneration by synthesizing nerve growth factor. Nat Cell Biol 2023; 25:1746-1757. [PMID: 38012403 PMCID: PMC10709146 DOI: 10.1038/s41556-023-01284-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 10/09/2023] [Indexed: 11/29/2023]
Abstract
The bone marrow contains peripheral nerves that promote haematopoietic regeneration after irradiation or chemotherapy (myeloablation), but little is known about how this is regulated. Here we found that nerve growth factor (NGF) produced by leptin receptor-expressing (LepR+) stromal cells is required to maintain nerve fibres in adult bone marrow. In nerveless bone marrow, steady-state haematopoiesis was normal but haematopoietic and vascular regeneration were impaired after myeloablation. LepR+ cells, and the adipocytes they gave rise to, increased NGF production after myeloablation, promoting nerve sprouting in the bone marrow and haematopoietic and vascular regeneration. Nerves promoted regeneration by activating β2 and β3 adrenergic receptor signalling in LepR+ cells, and potentially in adipocytes, increasing their production of multiple haematopoietic and vascular regeneration growth factors. Peripheral nerves and LepR+ cells thus promote bone marrow regeneration through a reciprocal relationship in which LepR+ cells sustain nerves by synthesizing NGF and nerves increase regeneration by promoting the production of growth factors by LepR+ cells.
Collapse
Affiliation(s)
- Xiang Gao
- National Institute of Biological Sciences, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Malea M Murphy
- Children's Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Integrated Microscopy and Imaging Laboratory, Texas A&M Health Science Center, Texas A&M University, College Station, TX, USA
| | - James G Peyer
- Children's Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cambrian Bio, Inc., New York, NY, USA
| | - Yuehan Ni
- National Institute of Biological Sciences, Beijing, China
- College of Life Sciences, Beijing Normal University, Beijing, China
| | - Min Yang
- National Institute of Biological Sciences, Beijing, China
- College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yixuan Zhang
- National Institute of Biological Sciences, Beijing, China
| | - Jiaming Guo
- National Institute of Biological Sciences, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Nergis Kara
- Children's Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Ensoma, Inc., Boston, MA, USA
| | - Claire Embree
- Children's Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alpaslan Tasdogan
- Children's Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Dermatology, University Hospital Essen and German Cancer Consortium, Essen, Germany
| | - Jessalyn M Ubellacker
- Children's Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Genevieve M Crane
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Shentong Fang
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhiyu Zhao
- Children's Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bo Shen
- National Institute of Biological Sciences, Beijing, China.
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China.
- Children's Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Sean J Morrison
- Children's Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Howard Hughes Medical Institute, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
49
|
Kasbekar M, Mitchell CA, Proven MA, Passegué E. Hematopoietic stem cells through the ages: A lifetime of adaptation to organismal demands. Cell Stem Cell 2023; 30:1403-1420. [PMID: 37865087 PMCID: PMC10842631 DOI: 10.1016/j.stem.2023.09.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/20/2023] [Accepted: 09/28/2023] [Indexed: 10/23/2023]
Abstract
Hematopoietic stem cells (HSCs), which govern the production of all blood lineages, transition through a series of functional states characterized by expansion during fetal development, functional quiescence in adulthood, and decline upon aging. We describe central features of HSC regulation during ontogeny to contextualize how adaptive responses over the life of the organism ultimately form the basis for HSC functional degradation with age. We particularly focus on the role of cell cycle regulation, inflammatory response pathways, epigenetic changes, and metabolic regulation. We then explore how the knowledge of age-related changes in HSC regulation can inform strategies for the rejuvenation of old HSCs.
Collapse
Affiliation(s)
- Monica Kasbekar
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University, New York, NY 10032, USA; Division of Hematology and Medical Oncology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Carl A Mitchell
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University, New York, NY 10032, USA
| | - Melissa A Proven
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University, New York, NY 10032, USA
| | - Emmanuelle Passegué
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
50
|
Wu J, Hu M, Jiang H, Ma J, Xie C, Zhang Z, Zhou X, Zhao J, Tao Z, Meng Y, Cai Z, Song T, Zhang C, Gao R, Cai C, Song H, Gao Y, Lin T, Wang C, Zhou X. Endothelial Cell-Derived Lactate Triggers Bone Mesenchymal Stem Cell Histone Lactylation to Attenuate Osteoporosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301300. [PMID: 37752768 PMCID: PMC10625121 DOI: 10.1002/advs.202301300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 08/17/2023] [Indexed: 09/28/2023]
Abstract
Blood vessels play a role in osteogenesis and osteoporosis; however, the role of vascular metabolism in these processes remains unclear. The present study finds that ovariectomized mice exhibit reduced blood vessel density in the bone and reduced expression of the endothelial glycolytic regulator pyruvate kinase M2 (PKM2). Endothelial cell (EC)-specific deletion of Pkm2 impairs osteogenesis and worsens osteoporosis in mice. This is attributed to the impaired ability of bone mesenchymal stem cells (BMSCs) to differentiate into osteoblasts. Mechanistically, EC-specific deletion of Pkm2 reduces serum lactate levels secreted by ECs, which affect histone lactylation in BMSCs. Using joint CUT&Tag and RNA sequencing analyses, collagen type I alpha 2 chain (COL1A2), cartilage oligomeric matrix protein (COMP), ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1), and transcription factor 7 like 2 (TCF7L2) as osteogenic genes regulated by histone H3K18la lactylation are identified. PKM2 overexpression in ECs, lactate addition, and exercise restore the phenotype of endothelial PKM2-deficient mice. Furthermore, serum metabolomics indicate that patients with osteoporosis have relatively low lactate levels. Additionally, histone lactylation and related osteogenic genes of BMSCs are downregulated in patients with osteoporosis. In conclusion, glycolysis in ECs fuels BMSC differentiation into osteoblasts through histone lactylation, and exercise partially ameliorates osteoporosis by increasing serum lactate levels.
Collapse
Affiliation(s)
- Jinhui Wu
- Department of OrthopedicsChangzheng HospitalNaval Medical UniversityShanghai200003P. R. China
| | - Miao Hu
- Department of OrthopedicsGeneral Hospital of Southern Theatre Command of PLAGuangzhou510010P. R. China
| | - Heng Jiang
- Department of OrthopedicsChangzheng HospitalNaval Medical UniversityShanghai200003P. R. China
| | - Jun Ma
- Department of OrthopedicsShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
| | - Chong Xie
- Department of NeurologyRenji HospitalShanghai Jiaotong University School of MedicineShanghai200127P. R. China
| | - Zheng Zhang
- Department of OrthopedicsChangzheng HospitalNaval Medical UniversityShanghai200003P. R. China
| | - Xin Zhou
- Department of OrthopedicsChangzheng HospitalNaval Medical UniversityShanghai200003P. R. China
- Department of OrthopedicsShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080P. R. China
| | - Jianquan Zhao
- Department of OrthopedicsChangzheng HospitalNaval Medical UniversityShanghai200003P. R. China
| | - Zhengbo Tao
- Department of OrthopedicsChangzheng HospitalNaval Medical UniversityShanghai200003P. R. China
| | - Yichen Meng
- Department of OrthopedicsChangzheng HospitalNaval Medical UniversityShanghai200003P. R. China
| | - Zhuyun Cai
- Department of OrthopedicsChangzheng HospitalNaval Medical UniversityShanghai200003P. R. China
| | - Tengfei Song
- Department of OrthopedicsChangzheng HospitalNaval Medical UniversityShanghai200003P. R. China
| | - Chenglin Zhang
- Department of OrthopedicsChangzheng HospitalNaval Medical UniversityShanghai200003P. R. China
| | - Rui Gao
- Department of OrthopedicsChangzheng HospitalNaval Medical UniversityShanghai200003P. R. China
| | - Chang Cai
- Department of OphthalmologyChanghai HospitalShanghai200433P. R. China
| | - Hongyuan Song
- Department of OphthalmologyChanghai HospitalShanghai200433P. R. China
| | - Yang Gao
- Senior Department of OrthopedicsThe Fourth Medical Center of PLA General HospitalBeijing100048P. R. China
| | - Tao Lin
- Department of OrthopedicsChangzheng HospitalNaval Medical UniversityShanghai200003P. R. China
| | - Ce Wang
- Department of OrthopedicsChangzheng HospitalNaval Medical UniversityShanghai200003P. R. China
| | - Xuhui Zhou
- Department of OrthopedicsChangzheng HospitalNaval Medical UniversityShanghai200003P. R. China
| |
Collapse
|