1
|
Liu H, Liu L, Rosen CJ. Bone Marrow Adipocytes as Novel Regulators of Metabolic Homeostasis: Clinical Consequences of Bone Marrow Adiposity. Curr Obes Rep 2025; 14:9. [PMID: 39808256 DOI: 10.1007/s13679-024-00594-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 01/16/2025]
Abstract
PURPOSE OF REVIEW Bone marrow adipose tissue is a distinctive fat depot located within the skeleton, with the potential to influence both local and systemic metabolic processes. Although significant strides have been made in understanding bone marrow adipose tissue over the past decade, many questions remain regarding their precise lineage and functional roles. RECENT FINDINGS Recent studies have highlighted bone marrow adipose tissue's involvement in continuous cross-talk with other organs and systems, exerting both endocrine and paracrine functions that play a crucial role in metabolic homeostasis, skeletal remodeling, hematopoiesis, and the progression of bone metastases. The advancement of imaging techniques, particularly cross-sectional imaging, has profoundly expanded our understanding of the complexities beyond the traditional view of bone marrow adipose tissue as an inert depot. Notably, marrow adipocytes are anatomically and functionally distinct from brown, beige, and classic white adipocytes. Emerging evidence suggests that bone marrow adipocytes, bone marrow adipose tissue originate from the differentiation of bone marrow mesenchymal stromal cells; however, they appear to be a heterogeneous population with varying metabolic profiles, lipid compositions, secretory properties, and functional responses depending on their specific location within the bone marrow. This review provides an up-to-date synthesis of current knowledge on bone marrow adipocytes, emphasizing the relationships between bone marrow adipogenesis and factors such as aging, osteoporosis, obesity, and bone marrow tumors or metastases, thereby elucidating the mechanisms underlying musculoskeletal pathophysiology.
Collapse
Affiliation(s)
- Hanghang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology &, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
- Maine Medical Center Research Institute, Maine Medical Center, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Linyi Liu
- Maine Medical Center Research Institute, Maine Medical Center, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Clifford J Rosen
- Maine Medical Center Research Institute, Maine Medical Center, 81 Research Drive, Scarborough, ME, 04074, USA.
| |
Collapse
|
2
|
Sánchez-Lanzas R, Jiménez-Pompa A, Ganuza M. The evolving hematopoietic niche during development. Front Mol Biosci 2024; 11:1488199. [PMID: 39417006 PMCID: PMC11480086 DOI: 10.3389/fmolb.2024.1488199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Mammalian hematopoietic stem cells (HSCs) emerge from the hemogenic endothelium in the major embryonic arteries. HSCs undergo a complex journey first migrating to the fetal liver (FL) and from there to the fetal bone marrow (FBM), where they mostly remain during adult life. In this process, a pool of adult HSCs is produced, which sustains lifelong hematopoiesis. Multiple cellular components support HSC maturation and expansion and modulate their response to environmental and developmental cues. While the adult HSC niche has been extensively studied over the last two decades, the niches present in the major embryonic arteries, FL, FBM and perinatal bone marrow (BM) are poorly described. Recent investigations highlight important differences among FL, FBM and adult BM niches and emphasize the important role that inflammation, microbiota and hormonal factors play regulating HSCs and their niches. We provide a review on our current understanding of these important cellular microenvironments across ontogeny. We mainly focused on mice, as the most widely used research model, and, when possible, include relevant insights from other vertebrates including birds, zebrafish, and human. Developing a comprehensive picture on these processes is critical to understand the earliest origins of childhood leukemia and to achieve multiple goals in regenerative medicine, such as mimicking HSC development in vitro to produce HSCs for broad transplantation purposes in leukemia, following chemotherapy, bone marrow failure, and in HSC-based gene therapy.
Collapse
Affiliation(s)
| | | | - Miguel Ganuza
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
3
|
Niemann T, Joneleit J, Storm J, Nacke T, Wähnert D, Kaltschmidt C, Vordemvenne T, Kaltschmidt B. Analyzing Sex-Specific Dimorphism in Human Skeletal Stem Cells. Cells 2023; 12:2683. [PMID: 38067111 PMCID: PMC10705359 DOI: 10.3390/cells12232683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/17/2023] [Accepted: 11/18/2023] [Indexed: 12/18/2023] Open
Abstract
Sex-related differences are a current topic in contemporary science. In addition to hormonal regulation, cell-autonomous mechanisms are important in bone homeostasis and regeneration. In this study, human skeletal stem cells (SSCs) from female and male adults were cultured and analyzed with immunological assays and osteogenic differentiation assessments. Female SSCs exhibited a mean doubling time of 100.6 h, whereas male SSCs displayed a mean doubling time of 168.0 h. Immunophenotyping revealed the expression of the stem cell markers Nestin, CD133, and CD164, accompanied by the neural-crest marker SOX9. Furthermore, multiparameter flow cytometric analyses revealed a substantial population of multipotent SSCs, comprising up to 80% in both sexes. An analysis of the osteogenic differentiation potential demonstrated a strong mineralization in both male and female SSCs under physiological conditions. Recognizing the prevailing association of bone diseases with inflammatory processes, we also analyzed the osteogenic potential of SSCs from both sexes under pro-inflammatory conditions. Upon TNF-α and IL-1β treatment, we observed no sexual dimorphism on osteogenesis. In summary, we demonstrated the successful isolation and characterization of SSCs capable of rapid osteogenic differentiation. Taken together, in vitro cultured SSCs might be a suitable model to study sexual dimorphisms and develop drugs for degenerative bone diseases.
Collapse
Affiliation(s)
- Tarek Niemann
- Molecular Neurobiology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany; (J.J.); (T.N.); (B.K.)
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., 33615 Bielefeld, Germany; (J.S.); (D.W.); (C.K.); (T.V.)
| | - Jonas Joneleit
- Molecular Neurobiology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany; (J.J.); (T.N.); (B.K.)
| | - Jonathan Storm
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., 33615 Bielefeld, Germany; (J.S.); (D.W.); (C.K.); (T.V.)
- Department of Cell Biology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany
| | - Tom Nacke
- Molecular Neurobiology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany; (J.J.); (T.N.); (B.K.)
| | - Dirk Wähnert
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., 33615 Bielefeld, Germany; (J.S.); (D.W.); (C.K.); (T.V.)
- Department of Trauma and Orthopedic Surgery, Protestant Hospital of Bethel Foundation, Campus Bielefeld-Bethel, University Hospital OWL of Bielefeld University, Burgsteig 13, 33617 Bielefeld, Germany
| | - Christian Kaltschmidt
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., 33615 Bielefeld, Germany; (J.S.); (D.W.); (C.K.); (T.V.)
- Department of Cell Biology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany
| | - Thomas Vordemvenne
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., 33615 Bielefeld, Germany; (J.S.); (D.W.); (C.K.); (T.V.)
- Department of Trauma and Orthopedic Surgery, Protestant Hospital of Bethel Foundation, Campus Bielefeld-Bethel, University Hospital OWL of Bielefeld University, Burgsteig 13, 33617 Bielefeld, Germany
| | - Barbara Kaltschmidt
- Molecular Neurobiology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany; (J.J.); (T.N.); (B.K.)
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., 33615 Bielefeld, Germany; (J.S.); (D.W.); (C.K.); (T.V.)
- Department of Cell Biology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany
| |
Collapse
|
4
|
Wu S, Ohba S, Matsushita Y. Single-Cell RNA-Sequencing Reveals the Skeletal Cellular Dynamics in Bone Repair and Osteoporosis. Int J Mol Sci 2023; 24:9814. [PMID: 37372962 DOI: 10.3390/ijms24129814] [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: 04/28/2023] [Revised: 05/29/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
The bone is an important organ that performs various functions, and the bone marrow inside the skeleton is composed of a complex intermix of hematopoietic, vascular, and skeletal cells. Current single-cell RNA sequencing (scRNA-seq) technology has revealed heterogeneity and sketchy differential hierarchy of skeletal cells. Skeletal stem and progenitor cells (SSPCs) are located upstream of the hierarchy and differentiate into chondrocytes, osteoblasts, osteocytes, and bone marrow adipocytes. In the bone marrow, multiple types of bone marrow stromal cells (BMSCs), which have the potential of SSPCs, are spatiotemporally located in distinct areas, and SSPCs' potential shift of BMSCs may occur with the advancement of age. These BMSCs contribute to bone regeneration and bone diseases, such as osteoporosis. In vivo lineage-tracing technologies show that various types of skeletal lineage cells concomitantly gather and contribute to bone regeneration. In contrast, these cells differentiate into adipocytes with aging, leading to senile osteoporosis. scRNA-seq analysis has revealed that alteration in the cell-type composition is a major cause of tissue aging. In this review, we discuss the cellular dynamics of skeletal cell populations in bone homeostasis, regeneration, and osteoporosis.
Collapse
Affiliation(s)
- Sixun Wu
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Shinsuke Ohba
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
- Department of Tissue and Developmental Biology, Osaka University Graduate School of Dentistry, Osaka 565-0871, Japan
| | - Yuki Matsushita
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| |
Collapse
|
5
|
O’Neill HC, Lim HK. Skeletal stem/progenitor cells provide the niche for extramedullary hematopoiesis in spleen. Front Physiol 2023; 14:1148414. [PMID: 37007998 PMCID: PMC10063897 DOI: 10.3389/fphys.2023.1148414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/10/2023] [Indexed: 03/19/2023] Open
Abstract
In bone marrow, the niche which supports hematopoiesis and nurtures hematopoietic stem cells (HSCs) contains perivascular reticular cells representing a subset of skeletal stem/progenitor cells (SSPCs). These stromal cells which provide the niche are lost or become inadequate during stress, disease or ageing, such that HSCs leave bone marrow and enter spleen and other peripheral sites to initiate extramedullary hematopoiesis and particularly myelopoiesis. Spleen also maintains niches for HSCs under steady-state conditions, evident since neonatal and adult spleen contain HSCs in low number and provide low-level hematopoiesis. In spleen, HSCs are found in the sinusoidal-rich red pulp region also in the vicinity of perivascular reticular cells. These cells resemble to some extent the known stromal elements reflecting HSC niches in bone marrow, and are investigated here for their characteristics as a subset of SSPCs. The isolation of spleen stromal subsets and the generation of cell lines which support HSCs and myelopoiesis in vitro has led to the identification of perivascular reticular cells which are unique to spleen. Analysis of gene and marker expression, as well as differentiative potential, identifies an osteoprogenitor cell type, reflective of one of several subsets of SSPCs described previously in bone, bone marrow and adipose tissue. The combined information supports a model for HSC niches in spleen involving perivascular reticular cells as SSPCs having osteogenic, stroma-forming capacity. These associate with sinusoids in red pulp to form niches for HSCs and to support the differentiation of hematopoietic progenitors during extramedullary hematopoiesis.
Collapse
|
6
|
Yuan G, Li Z, Lin X, Li N, Xu R. New perspective of skeletal stem cells. BIOMATERIALS TRANSLATIONAL 2022; 3:280-294. [PMID: 36846511 PMCID: PMC9947737 DOI: 10.12336/biomatertransl.2022.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 11/29/2022] [Accepted: 12/19/2022] [Indexed: 03/01/2023]
Abstract
Tissue-resident stem cells are a group of stem cells distinguished by their capacity for self-renewal and multilineage differentiation capability with tissue specificity. Among these tissue-resident stem cells, skeletal stem cells (SSCs) were discovered in the growth plate region through a combination of cell surface markers and lineage tracing series. With the process of unravelling the anatomical variation of SSCs, researchers were also keen to investigate the developmental diversity outside the long bones, including in the sutures, craniofacial sites, and spinal regions. Recently, fluorescence-activated cell sorting, lineage tracing, and single-cell sequencing have been used to map lineage trajectories by studying SSCs with different spatiotemporal distributions. The SSC niche also plays a pivotal role in regulating SSC fate, such as cell-cell interactions mediated by multiple signalling pathways. This review focuses on discussing the spatial and temporal distribution of SSCs, and broadening our understanding of the diversity and plasticity of SSCs by summarizing the progress of research into SSCs in recent years.
Collapse
Affiliation(s)
- Guixin Yuan
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Centre for Skeletal Stem Cell, State Key Laboratory of Cellular Stress Biology, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian Province, China,Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, School of Medicine, Xiamen University, Xiamen, Fujian Province, China,Department of Human Anatomy, School of Medicine, Xiamen University, Xiamen, Fujian Province, China
| | - Zan Li
- Department of Sports Medicine & Research Centre of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Xixi Lin
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Centre for Skeletal Stem Cell, State Key Laboratory of Cellular Stress Biology, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian Province, China,Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, School of Medicine, Xiamen University, Xiamen, Fujian Province, China,Department of Human Anatomy, School of Medicine, Xiamen University, Xiamen, Fujian Province, China
| | - Na Li
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Centre for Skeletal Stem Cell, State Key Laboratory of Cellular Stress Biology, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian Province, China,Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, School of Medicine, Xiamen University, Xiamen, Fujian Province, China,Department of Human Anatomy, School of Medicine, Xiamen University, Xiamen, Fujian Province, China,Corresponding authors: Ren Xu, ; Na Li,
| | - Ren Xu
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Centre for Skeletal Stem Cell, State Key Laboratory of Cellular Stress Biology, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian Province, China,Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, School of Medicine, Xiamen University, Xiamen, Fujian Province, China,Department of Human Anatomy, School of Medicine, Xiamen University, Xiamen, Fujian Province, China,Corresponding authors: Ren Xu, ; Na Li,
| |
Collapse
|
7
|
Tsukagoshi Y, Matsushita Y. Bone regeneration: A message from clinical medicine and basic science. Clin Anat 2022; 35:808-819. [PMID: 35654609 DOI: 10.1002/ca.23917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 05/27/2022] [Indexed: 11/08/2022]
Abstract
Population aging is a global phenomenon and with it, the number of bone fractures increases due to higher incidences of osteoporosis. Bone fractures in the elderly increase the risk of bedridden status and mortality. Therefore, the control of osteoporosis and bone fracture is important for healthy life expectancy, and the fundamental understanding of its pathogenesis and its application in treatment is of great social significance. To solve these clinical problems, it is necessary to integrate clinical medicine and basic research. Bone regeneration after a fracture is an essential function of the living body. The prevailing view is that a small number of resident skeletal stem cells are solely responsible for regenerative capacity. Although these cells have long been considered to be in the bone marrow, it has been shown that they are also present in the growth plate and periosteum. More recently, distinct types of cells in the bone marrow, including bone marrow stromal cells, osteoblast progenitor cells, and osteoblasts, have been shown to participate in bone regeneration. Interestingly, the cellular plasticity of differentiated cells, rather than active recruitment of resident stem cell populations, may largely account for regeneration of bone tissues; terminally differentiated cells de-differentiate into a stem cell-like state, and then re-differentiate into regenerating bone. In this review, we discuss the clinical risk and preventive therapy of bone fractures and the current concept of bone regeneration in basic mechanical insights, which may prove useful to both clinicians and researchers.
Collapse
Affiliation(s)
- Yuta Tsukagoshi
- Department of Orthopedic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yuki Matsushita
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
8
|
Li W, Li T, Tang Z, Qi X, Zhou Y, Tang X, Xu W, Xiong H. Taohong Siwu Decoction promotes the process of fracture healing by activating the VEGF-FAK signal pathway and systemically regulating the gut microbiota. J Appl Microbiol 2022; 133:1363-1377. [PMID: 35475538 DOI: 10.1111/jam.15598] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 04/07/2022] [Accepted: 04/25/2022] [Indexed: 11/27/2022]
Abstract
AIMS This study aimed to explore the effect of Taohong Siwu Decoction (THSWD) on Bone marrow mesenchymal stem cells (BMSCs) at the cellular level and the possible mechanism of systemic regulation of gut microbiota on fracture recovery. METHODS AND RESULTS Cell Counting Kit-8 (CCK-8) experiments show that THSWD effectively promotes the proliferation of BMSCs. Transwell and wound healing assays show that THSWD effectively promotes the invasion and migration of BMSCs. Alizarin red staining showed that the THSWD model enhanced the osteogenic differentiation of BMSCs. Moreover, the effect of THSWD on BMSCs is time- and concentration-dependent. RT-qPCR and Western blot results showed that THSWD treatment up-regulated the expression of vascular endothelial growth factor (VEGF) and focal adhesion kinase (FAK) at mRNA and protein levels, respectively. Hematoxylin-eosin and crocin O-quick green staining showed that rats with right femoral shaft fractures, after 14 days of THSWD treatment, the area of callus and cartilage regeneration at the fracture site increased significantly. Gut microbiota was changed in fractured rats, such as the abundance of Bacteroidetes and Firmicutes was increased. THSWD showed positive regulation of both to a certain extent. CONCLUSION THSWD up-regulates VEGF and activates the FAK signaling pathway to enhance the development and differentiation of BMSCs, and systematically regulates the gut microbiota to promote fracture healing. SIGNIFICANCE AND IMPACT OF STUDY This study provides new insights on the cellular and systemic level to understand the mechanism of THSWD in the treatment of fractures.
Collapse
Affiliation(s)
- Wangyang Li
- Hunan University of Chinese Medicine, Changsha, Hunan
| | - Tiao Li
- Department of Respiratory Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan
| | - Zhi Tang
- Xiangtan Chinese Medicine hospital, Xiangtan, Hunan
| | - Xinyu Qi
- Hunan University of Chinese Medicine, Changsha, Hunan
| | - Youliang Zhou
- Department of Emergency First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, Hunan
| | - Xiaolu Tang
- Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan
| | - Weijie Xu
- Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan
| | - Hui Xiong
- Hunan University of Chinese Medicine, Changsha, Hunan
| |
Collapse
|
9
|
Matsushita Y, Ono W, Ono N. Toward Marrow Adipocytes: Adipogenic Trajectory of the Bone Marrow Stromal Cell Lineage. Front Endocrinol (Lausanne) 2022; 13:882297. [PMID: 35528017 PMCID: PMC9075612 DOI: 10.3389/fendo.2022.882297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 03/24/2022] [Indexed: 11/13/2022] Open
Abstract
Bone marrow contains precursor cells for osteoblasts and adipocytes in the stromal compartment. Bone marrow adipose tissue (BMAT) is an important constituent of the bone marrow that is particularly abundant in adults. BMAT is composed of the proximal "regulated" BMAT containing individual adipocytes interspersed within actively hematopoietic marrow, and the distal "constitutive" BMAT containing large adipocytes in the area of low hematopoiesis. Historically, bone marrow adipocytes were regarded as one of the terminal states of skeletal stem cells, which stand at the pinnacle of the lineage and possess trilineage differentiation potential into osteoblasts, chondrocytes and adipocytes. Recent single-cell RNA-sequencing studies uncover a discrete group of preadipocyte-like cells among bone marrow stromal cells (BMSCs), and recent mouse genetic lineage-tracing studies reveal that these adipocyte precursor cells possess diverse functions in homeostasis and regeneration. These adipogenic subsets of BMSCs are abundant in the central marrow space and can directly convert not only into lipid-laden adipocytes but also into skeletal stem cell-like cells and osteoblasts under regenerative conditions. It remains determined whether there are distinct adipocyte precursor cell types contributing to two types of BMATs. In this short review, we discuss the functions of the recently identified subsets of BMSCs and their trajectory toward marrow adipocytes, which is influenced by multiple modes of cell-autonomous and non-cell autonomous regulations.
Collapse
|
10
|
Tang YJ, Puviindran V, Xiang Y, Yahara Y, Zhang H, Nadesan P, Diao Y, Kirsch DG, Alman BA. Tumor-propagating side population cells are a dynamic subpopulation in undifferentiated pleomorphic sarcoma. JCI Insight 2021; 6:148768. [PMID: 34618689 PMCID: PMC8663789 DOI: 10.1172/jci.insight.148768] [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: 03/11/2021] [Accepted: 10/06/2021] [Indexed: 11/25/2022] Open
Abstract
Sarcomas contain a subpopulation of tumor-propagating cells (TPCs) with enhanced tumor-initiating and self-renewal properties. However, it is unclear whether the TPC phenotype in sarcomas is stable or a dynamic cell state that can derive from non-TPCs. In this study, we utilized a mouse model of undifferentiated pleomorphic sarcoma (UPS) to trace the lineage relationship between sarcoma side population (SP) cells that are enriched for TPCs and non-SP cells. By cotransplanting SP and non-SP cells expressing different endogenous fluorescent reporters, we show that non-SP cells can give rise to SP cells with enhanced tumor-propagating potential in vivo. Lineage trajectory analysis using single-cell RNA sequencing from SP and non-SP cells supports the notion that non-SP cells can assume the SP cell phenotype de novo. To test the effect of eradicating SP cells on tumor growth and self-renewal, we generated mouse sarcomas in which the diphtheria toxin receptor is expressed in the SP cells and their progeny. Ablation of the SP population using diphtheria toxin did not impede tumor growth or self-renewal. Altogether, we show that the sarcoma SP represent a dynamic cell state and targeting TPCs alone is insufficient to eliminate tumor progression.
Collapse
Affiliation(s)
- Yuning Jackie Tang
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Department of Orthopedic Surgery
| | | | - Yu Xiang
- Department of Cell Biology.,Regeneration Next Initiative
| | | | - Hongyuan Zhang
- Department of Orthopedic Surgery.,Department of Cell Biology
| | | | - Yarui Diao
- Department of Orthopedic Surgery.,Department of Cell Biology.,Regeneration Next Initiative
| | - David G Kirsch
- Regeneration Next Initiative.,Department of Pharmacology and Cancer Biology, and.,Department of Radiation Oncology, Duke University School of Medicine, Duke University, Durham, North Carolina, USA
| | - Benjamin A Alman
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Department of Orthopedic Surgery.,Department of Cell Biology.,Regeneration Next Initiative
| |
Collapse
|
11
|
Mizoguchi T, Ono N. The diverse origin of bone-forming osteoblasts. J Bone Miner Res 2021; 36:1432-1447. [PMID: 34213032 PMCID: PMC8338797 DOI: 10.1002/jbmr.4410] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 06/29/2021] [Accepted: 06/29/2021] [Indexed: 12/17/2022]
Abstract
Osteoblasts are the only cells that can give rise to bones in vertebrates. Thus, one of the most important functions of these metabolically active cells is mineralized matrix production. Because osteoblasts have a limited lifespan, they must be constantly replenished by preosteoblasts, their immediate precursors. Because disruption of the regulation of bone-forming osteoblasts results in a variety of bone diseases, a better understanding of the origin of these cells by defining the mechanisms of bone development, remodeling, and regeneration is central to the development of novel therapeutic approaches. In recent years, substantial new insights into the origin of osteoblasts-largely owing to rapid technological advances in murine lineage-tracing approaches and other single-cell technologies-have been obtained. Collectively, these findings indicate that osteoblasts involved in bone formation under various physiological, pathological, and therapeutic conditions can be obtained from numerous sources. The origins of osteoblasts include, but are not limited to, chondrocytes in the growth plate, stromal cells in the bone marrow, quiescent bone-lining cells on the bone surface, and specialized fibroblasts in the craniofacial structures, such as sutures and periodontal ligaments. Because osteoblasts can be generated from local cellular sources, bones can flexibly respond to regenerative and anabolic cues. However, whether osteoblasts derived from different cellular sources have distinct functions remains to be investigated. Currently, we are at the initial stage to aptly unravel the incredible diversity of the origins of bone-forming osteoblasts. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
| | - Noriaki Ono
- University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, USA
| |
Collapse
|
12
|
Ortinau LC, Park D. Do adipogenic stromal cells undergo lineage plasticity in response to bone injury? Bioessays 2020; 43:e2000296. [PMID: 33616991 DOI: 10.1002/bies.202000296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 11/11/2022]
Affiliation(s)
- Laura C Ortinau
- Department of Molecular Human Genetics, Center for Skeletal Biology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, USA
| | - Dongsu Park
- Department of Molecular Human Genetics, Center for Skeletal Biology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, USA
| |
Collapse
|