1
|
Weldon KC, Longaker MT, Ambrosi TH. Harnessing the diversity and potential of endogenous skeletal stem cells for musculoskeletal tissue regeneration. Stem Cells 2025; 43:sxaf006. [PMID: 39945760 PMCID: PMC11892563 DOI: 10.1093/stmcls/sxaf006] [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: 10/29/2024] [Accepted: 01/21/2025] [Indexed: 03/11/2025]
Abstract
In our aging society, the degeneration of the musculoskeletal system and adjacent tissues is a growing orthopedic concern. As bones age, they become more fragile, increasing the risk of fractures and injuries. Furthermore, tissues like cartilage accumulate damage, leading to widespread joint issues. Compounding this, the regenerative capacity of these tissues declines with age, exacerbating the consequences of fractures and cartilage deterioration. With rising demand for fracture and cartilage repair, bone-derived stem cells have attracted significant research interest. However, the therapeutic use of stem cells has produced inconsistent results, largely due to ongoing debates and uncertainties regarding the precise identity of the stem cells responsible for musculoskeletal growth, maintenance and repair. This review focuses on the potential to leverage endogenous skeletal stem cells (SSCs)-a well-defined population of stem cells with specific markers, reliable isolation techniques, and functional properties-in bone repair and cartilage regeneration. Understanding SSC behavior in response to injury, including their activation to a functional state, could provide insights into improving treatment outcomes. Techniques like microfracture surgery, which aim to stimulate SSC activity for cartilage repair, are of particular interest. Here, we explore the latest advances in how such interventions may modulate SSC function to enhance bone healing and cartilage regeneration.
Collapse
Affiliation(s)
- Kelly C Weldon
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA 95817, United States
- School of Medicine, University of California, Sacramento, CA 95817, United States
| | - Michael T Longaker
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, United States
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, United States
| | - Thomas H Ambrosi
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA 95817, United States
| |
Collapse
|
2
|
Cong T, Morse KW, Sosa BR, Lane JM, Rodeo SA, Greenblatt MB. Skeletal Stem Cells: A Basis for Orthopaedic Pathology and Tissue Repair. J Bone Joint Surg Am 2025; 107:418-426. [PMID: 39693451 PMCID: PMC11839314 DOI: 10.2106/jbjs.24.00905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
➢ Skeletal stem cells (SSCs) continually replenish mature cell populations to support skeletal homeostasis.➢ SSCs repopulate by self-renewal, have multilineage potential, and are long-lived in vivo.➢ SSCs express specific combinations of cell surface markers that reflect their lineage identity.➢ SSCs adapt to their anatomic environment to support regional differences in skeletal behavior and pathology.
Collapse
Affiliation(s)
- Ting Cong
- Department of Orthopaedic Surgery, UPMC Sports Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
- Department of Orthopedic Surgery, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| | - Kyle W Morse
- Hospital for Special Surgery, New York, NY
- Department of Orthopaedic Surgery, Weill Cornell Medicine, New York, NY
| | - Branden R Sosa
- Hospital for Special Surgery, New York, NY
- Department of Orthopaedic Surgery, Weill Cornell Medicine, New York, NY
| | - Joseph M Lane
- Hospital for Special Surgery, New York, NY
- Department of Orthopaedic Surgery, Weill Cornell Medicine, New York, NY
| | - Scott A Rodeo
- Hospital for Special Surgery, New York, NY
- Department of Orthopaedic Surgery, Weill Cornell Medicine, New York, NY
| | - Matthew B Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
- Research Division, Hospital for Special Surgery, New York, NY
| |
Collapse
|
3
|
Reeves J, Tournier P, Becquart P, Carton R, Tang Y, Vigilante A, Fang D, Habib SJ. Rejuvenating aged osteoprogenitors for bone repair. eLife 2024; 13:RP104068. [PMID: 39692737 DOI: 10.7554/elife.104068] [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] [Indexed: 12/19/2024] Open
Abstract
Aging is marked by a decline in tissue regeneration, posing significant challenges to an increasingly older population. Here, we investigate age-related impairments in calvarial bone healing and introduce a novel two-part rejuvenation strategy to restore youthful repair. We demonstrate that aging negatively impacts the calvarial bone structure and its osteogenic tissues, diminishing osteoprogenitor number and function and severely impairing bone formation. Notably, increasing osteogenic cell numbers locally fails to rescue repair in aged mice, identifying the presence of intrinsic cellular deficits. Our strategy combines Wnt-mediated osteoprogenitor expansion with intermittent fasting, which leads to a striking restoration of youthful levels of bone healing. We find that intermittent fasting improves osteoprogenitor function, benefits that can be recapitulated by modulating NAD+-dependent pathways or the gut microbiota, underscoring the multifaceted nature of this intervention. Mechanistically, we identify mitochondrial dysfunction as a key component in age-related decline in osteoprogenitor function and show that both cyclical nutrient deprivation and Nicotinamide mononucleotide rejuvenate mitochondrial health, enhancing osteogenesis. These findings offer a promising therapeutic avenue for restoring youthful bone repair in aged individuals, with potential implications for rejuvenating other tissues.
Collapse
Affiliation(s)
- Joshua Reeves
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
- Centre for Gene Therapy and Regenerative Medicine King's College London, London, United Kingdom
| | - Pierre Tournier
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Pierre Becquart
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Robert Carton
- Centre for Gene Therapy and Regenerative Medicine King's College London, London, United Kingdom
| | - Yin Tang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute Zhejiang University, Zhejiang, China
- Department of Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Alessandra Vigilante
- Centre for Gene Therapy and Regenerative Medicine King's College London, London, United Kingdom
| | - Dong Fang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute Zhejiang University, Zhejiang, China
- Department of Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Shukry J Habib
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
4
|
Liang J, Wang J, Sui B, Tong Y, Chai J, Zhou Q, Zheng C, Wang H, Kong L, Zhang H, Bai Y. Ptip safeguards the epigenetic control of skeletal stem cell quiescence and potency in skeletogenesis. Sci Bull (Beijing) 2024; 69:2099-2113. [PMID: 38493069 DOI: 10.1016/j.scib.2024.02.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/23/2023] [Accepted: 02/21/2024] [Indexed: 03/18/2024]
Abstract
Stem cells remain in a quiescent state for long-term maintenance and preservation of potency; this process requires fine-tuning regulatory mechanisms. In this study, we identified the epigenetic landscape along the developmental trajectory of skeletal stem cells (SSCs) in skeletogenesis governed by a key regulator, Ptip (also known as Paxip1, Pax interaction with transcription-activation domain protein-1). Our results showed that Ptip is required for maintaining the quiescence and potency of SSCs, and loss of Ptip in type II collagen (Col2)+ progenitors causes abnormal activation and differentiation of SSCs, impaired growth plate morphogenesis, and long bone dysplasia. We also found that Ptip suppressed the glycolysis of SSCs through downregulation of phosphoglycerate kinase 1 (Pgk1) by repressing histone H3 lysine 27 acetylation (H3K27ac) at the promoter region. Notably, inhibition of glycolysis improved the function of SSCs despite Ptip deficiency. To the best of our knowledge, this is the first study to establish an epigenetic framework based on Ptip, which safeguards skeletal stem cell quiescence and potency through metabolic control. This framework is expected to improve SSC-based treatments of bone developmental disorders.
Collapse
Affiliation(s)
- Jianfei Liang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China; Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China; Department of Implant Dentistry, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China; State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Jing Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
| | - Bingdong Sui
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Yibo Tong
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Jihua Chai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
| | - Qin Zhou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China; Department of Implant Dentistry, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China
| | - Chenxi Zheng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Hao Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China
| | - Liang Kong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, China.
| | - Haojian Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China; Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan 430079, China; Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan 430079, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China.
| | - Yi Bai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China.
| |
Collapse
|
5
|
Patrick R, Janbandhu V, Tallapragada V, Tan SSM, McKinna EE, Contreras O, Ghazanfar S, Humphreys DT, Murray NJ, Tran YTH, Hume RD, Chong JJH, Harvey RP. Integration mapping of cardiac fibroblast single-cell transcriptomes elucidates cellular principles of fibrosis in diverse pathologies. SCIENCE ADVANCES 2024; 10:eadk8501. [PMID: 38905342 PMCID: PMC11192082 DOI: 10.1126/sciadv.adk8501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 05/14/2024] [Indexed: 06/23/2024]
Abstract
Single-cell technology has allowed researchers to probe tissue complexity and dynamics at unprecedented depth in health and disease. However, the generation of high-dimensionality single-cell atlases and virtual three-dimensional tissues requires integrated reference maps that harmonize disparate experimental designs, analytical pipelines, and taxonomies. Here, we present a comprehensive single-cell transcriptome integration map of cardiac fibrosis, which underpins pathophysiology in most cardiovascular diseases. Our findings reveal similarity between cardiac fibroblast (CF) identities and dynamics in ischemic versus pressure overload models of cardiomyopathy. We also describe timelines for commitment of activated CFs to proliferation and myofibrogenesis, profibrotic and antifibrotic polarization of myofibroblasts and matrifibrocytes, and CF conservation across mouse and human healthy and diseased hearts. These insights have the potential to inform knowledge-based therapies.
Collapse
Affiliation(s)
- Ralph Patrick
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Vaibhao Janbandhu
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
| | | | - Shannon S. M. Tan
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Emily E. McKinna
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia
| | - Osvaldo Contreras
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Shila Ghazanfar
- School of Mathematics and Statistics, The University of Sydney, Camperdown, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- Sydney Precision Data Science Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| | - David T. Humphreys
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Nicholas J. Murray
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Yen T. H. Tran
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Robert D. Hume
- Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia
- School of Medical Science, The University of Sydney, Camperdown, NSW 2006, Australia
- Centre for Heart Failure and Diseases of the Aorta, The Baird Institute, Sydney, NSW 2042, Australia
| | - James J. H. Chong
- Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia
- Department of Cardiology, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Richard P. Harvey
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
- School of Biotechnology and Biomolecular Science, UNSW Sydney, Kensington, NSW 2052, Australia
| |
Collapse
|
6
|
Sayed IM, Vo DT, Alcantara J, Inouye KM, Pranadinata RF, Luo L, Boland CR, Goyal NP, Kuo DJ, Huang SC, Sahoo D, Ghosh P, Das S. Molecular Signatures for Microbe-Associated Colorectal Cancers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.26.595902. [PMID: 38853996 PMCID: PMC11160670 DOI: 10.1101/2024.05.26.595902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Background Genetic factors and microbial imbalances play crucial roles in colorectal cancers (CRCs), yet the impact of infections on cancer initiation remains poorly understood. While bioinformatic approaches offer valuable insights, the rising incidence of CRCs creates a pressing need to precisely identify early CRC events. We constructed a network model to identify continuum states during CRC initiation spanning normal colonic tissue to pre-cancer lesions (adenomatous polyps) and examined the influence of microbes and host genetics. Methods A Boolean network was built using a publicly available transcriptomic dataset from healthy and adenoma affected patients to identify an invariant Microbe-Associated Colorectal Cancer Signature (MACS). We focused on Fusobacterium nucleatum ( Fn ), a CRC-associated microbe, as a model bacterium. MACS-associated genes and proteins were validated by RT-qPCR, RNA seq, ELISA, IF and IHCs in tissues and colon-derived organoids from genetically predisposed mice ( CPC-APC Min+/- ) and patients (FAP, Lynch Syndrome, PJS, and JPS). Results The MACS that is upregulated in adenomas consists of four core genes/proteins: CLDN2/Claudin-2 (leakiness), LGR5/leucine-rich repeat-containing receptor (stemness), CEMIP/cell migration-inducing and hyaluronan-binding protein (epithelial-mesenchymal transition) and IL8/Interleukin-8 (inflammation). MACS was induced upon Fn infection, but not in response to infection with other enteric bacteria or probiotics. MACS induction upon Fn infection was higher in CPC-APC Min+/- organoids compared to WT controls. The degree of MACS expression in the patient-derived organoids (PDOs) generally corresponded with the known lifetime risk of CRCs. Conclusions Computational prediction followed by validation in the organoid-based disease model identified the early events in CRC initiation. MACS reveals that the CRC-associated microbes induce a greater risk in the genetically predisposed hosts, suggesting its potential use for risk prediction and targeted cancer prevention.
Collapse
|
7
|
Saul D, Doolittle ML, Rowsey JL, Froemming MN, Kosinsky RL, Vos SJ, Ruan M, LeBrasseur NK, Chandra A, Pignolo RJ, Passos JF, Farr JN, Monroe DG, Khosla S. Osteochondroprogenitor cells and neutrophils expressing p21 and senescence markers modulate fracture repair. J Clin Invest 2024; 134:e179834. [PMID: 38753433 PMCID: PMC11178538 DOI: 10.1172/jci179834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 05/01/2024] [Indexed: 05/18/2024] Open
Abstract
Cells expressing features of senescence, including upregulation of p21 and p16, appear transiently following tissue injury, yet the properties of these cells or how they contrast with age-induced senescent cells remains unclear. Here, we used skeletal injury as a model and identified the rapid appearance following fracture of p21+ cells expressing senescence markers, mainly as osteochondroprogenitors (OCHs) and neutrophils. Targeted genetic clearance of p21+ cells suppressed senescence-associated signatures within the fracture callus and accelerated fracture healing. By contrast, p21+ cell clearance did not alter bone loss due to aging; conversely, p16+ cell clearance, known to alleviate skeletal aging, did not affect fracture healing. Following fracture, p21+ neutrophils were enriched in signaling pathways known to induce paracrine stromal senescence, while p21+ OCHs were highly enriched in senescence-associated secretory phenotype factors known to impair bone formation. Further analysis revealed an injury-specific stem cell-like OCH subset that was p21+ and highly inflammatory, with a similar inflammatory mesenchymal population (fibro-adipogenic progenitors) evident following muscle injury. Thus, intercommunicating senescent-like neutrophils and mesenchymal progenitor cells were key regulators of tissue repair in bone and potentially across tissues. Moreover, our findings established contextual roles of p21+ versus p16+ senescent/senescent-like cells that may be leveraged for therapeutic opportunities.
Collapse
Affiliation(s)
- Dominik Saul
- Division of Endocrinology and
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
- Department of Trauma and Reconstructive Surgery, BG Clinic, University of Tübingen, Tübingen, Germany
| | - Madison L. Doolittle
- Division of Endocrinology and
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | - Jennifer L. Rowsey
- Division of Endocrinology and
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | - Mitchell N. Froemming
- Division of Endocrinology and
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | - Robyn L. Kosinsky
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
- Robert Bosch Center for Tumor Diseases, Stuttgart, Germany
| | - Stephanie J. Vos
- Division of Endocrinology and
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | - Ming Ruan
- Division of Endocrinology and
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | - Nathan K. LeBrasseur
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physical Medicine and Rehabilitation and
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Abhishek Chandra
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Robert J. Pignolo
- Division of Endocrinology and
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - João F. Passos
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Joshua N. Farr
- Division of Endocrinology and
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - David G. Monroe
- Division of Endocrinology and
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | - Sundeep Khosla
- Division of Endocrinology and
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
8
|
Saul D, Doolittle ML, Rowsey JL, Froemming MN, Kosinsky RL, Vos SJ, Ruan M, LeBrasseur N, Chandra A, Pignolo R, Passos JF, Farr JN, Monroe DG, Khosla S. Osteochondroprogenitor cells and neutrophils expressing p21 and senescence markers modulate fracture repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.01.578420. [PMID: 38370844 PMCID: PMC10871229 DOI: 10.1101/2024.02.01.578420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Cells expressing features of senescence, including upregulation of p21 and p16, appear transiently following tissue injury, yet the properties of these cells or how they contrast with age-induced senescent cells remains unclear. Here, we used skeletal injury as a model and identified the rapid appearance following fracture of p21+ cells expressing senescence markers, mainly as osteochondroprogenitors (OCHs) and neutrophils. Targeted genetic clearance of p21+ cells suppressed senescence-associated signatures within the fracture callus and accelerated fracture healing. By contrast, p21+ cell clearance did not alter bone loss due to aging; conversely, p16+ cell clearance, known to alleviate skeletal aging, did not affect fracture healing. Following fracture, p21+ neutrophils were enriched in signaling pathways known to induce paracrine stromal senescence, while p21+ OCHs were highly enriched in senescence-associated secretory phenotype factors known to impair bone formation. Further analysis revealed an injury-specific stem cell-like OCH subset that was p21+ and highly inflammatory, with a similar inflammatory mesenchymal population (fibro-adipogenic progenitors) evident following muscle injury. Thus, intercommunicating senescent-like neutrophils and mesenchymal progenitor cells are key regulators of tissue repair in bone and potentially across tissues. Moreover, our findings establish contextual roles of p21+ vs p16+ senescent/senescent-like cells that may be leveraged for therapeutic opportunities.
Collapse
|
9
|
Xining Z, Sai L. The Evolving Function of Vasculature and Pro-angiogenic Therapy in Fat Grafting. Cell Transplant 2024; 33:9636897241264976. [PMID: 39056562 PMCID: PMC11282510 DOI: 10.1177/09636897241264976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 07/28/2024] Open
Abstract
Autologous fat grating is a widely-accepted method to correct soft tissue deficiency. Although fat transplantation shows excellent biocompatibility and simple applicability, the relatively low retention rate caused by fat necrosis is still a challenge. The vasculature is integral after fat grafting, serving multiple crucial functions. Rapid and effective angiogenesis within grafts is essential for supplying oxygen necessary for adipocytes' survival. It facilitates the influx of inflammatory cells to remove necrotic adipocytes and aids in the delivery of regenerative cells for adipose tissue regeneration in fat grafts. The vasculature also provides a niche for interaction between adipose progenitor cells and vascular progenitor cells, enhancing angiogenesis and adipogenesis in grafts. Various methods, such as enriching grafts with diverse pro-angiogenic cells or utilizing cell-free approaches, have been employed to enhance angiogenesis. Beige and dedifferentiated adipocytes in grafts could increase vessel density. This review aims to outline the function of vasculature in fat grafting and discuss different cell or cell-free approaches that can enhance angiogenesis following fat grafting.
Collapse
Affiliation(s)
- Zhang Xining
- The Plastic and Aesthetic Center, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Luo Sai
- The Plastic and Aesthetic Center, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
10
|
Qing J, Guo Q, Lv L, Zhang X, Liu Y, Heng BC, Li Z, Zhang P, Zhou Y. Organoid Culture Development for Skeletal Systems. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:545-557. [PMID: 37183418 DOI: 10.1089/ten.teb.2023.0022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Organoids are widely considered to be ideal in vitro models that have been widely applied in many fields, including regenerative medicine, disease research and drug screening. It is distinguished from other three-dimensional in vitro culture model systems by self-organization and sustainability in long-term culture. The three core components of organoid culture are cells, exogenous factors, and culture matrix. Due to the complexity of bone tissue, and heterogeneity of osteogenic stem/progenitor cells, it is challenging to construct organoids for modeling skeletal systems. In this study, we examine current progress in the development of skeletal system organoid culture systems and analyze the current research status of skeletal stem cells, their microenvironmental factors, and various potential organoid culture matrix candidates to provide cues for future research trajectory in this field. Impact Statement The emergence of organoids has brought new opportunities for the development of many biomedical fields. The bone organoid field still has much room for exploration. This review discusses the characteristics distinguishing organoids from other three-dimensional model systems and examines current progress in the organoid production of skeletal systems. In addition, based on core elements of organoid cultures, three main problems that need to be solved in bone organoid generation are further analyzed. These include the heterogeneity of skeletal stem cells, their microenvironmental factors, and potential organoid culture matrix candidates. This information provides direction for the future research of bone organoids.
Collapse
Affiliation(s)
- Jia Qing
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Qian Guo
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Longwei Lv
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Xiao Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Boon Chin Heng
- The Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
| | - Zheng Li
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Ping Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Haidian District, Beijing, China
| |
Collapse
|
11
|
Fiévet L, Espagnolle N, Gerovska D, Bernard D, Syrykh C, Laurent C, Layrolle P, De Lima J, Justo A, Reina N, Casteilla L, Araúzo-Bravo MJ, Naji A, Pagès JC, Deschaseaux F. Single-cell RNA sequencing of human non-hematopoietic bone marrow cells reveals a unique set of inter-species conserved biomarkers for native mesenchymal stromal cells. Stem Cell Res Ther 2023; 14:229. [PMID: 37649081 PMCID: PMC10469496 DOI: 10.1186/s13287-023-03437-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 07/28/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Native bone marrow (BM) mesenchymal stem/stromal cells (BM-MSCs) participate in generating and shaping the skeleton and BM throughout the lifespan. Moreover, BM-MSCs regulate hematopoiesis by contributing to the hematopoietic stem cell niche in providing critical cytokines, chemokines and extracellular matrix components. However, BM-MSCs contain a heterogeneous cell population that remains ill-defined. Although studies on the taxonomy of native BM-MSCs in mice have just started to emerge, the taxonomy of native human BM-MSCs remains unelucidated. METHODS By using single-cell RNA sequencing (scRNA-seq), we aimed to define a proper taxonomy for native human BM non-hematopoietic subsets including endothelial cells (ECs) and mural cells (MCs) but with a focal point on MSCs. To this end, transcriptomic scRNA-seq data were generated from 5 distinct BM donors and were analyzed together with other transcriptomic data and with computational biology analyses at different levels to identify, characterize and classify distinct native cell subsets with relevant biomarkers. RESULTS We could ascribe novel specific biomarkers to ECs, MCs and MSCs. Unlike ECs and MCs, MSCs exhibited an adipogenic transcriptomic pattern while co-expressing genes related to hematopoiesis support and multilineage commitment potential. Furthermore, by a comparative analysis of scRNA-seq of BM cells from humans and mice, we identified core genes conserved in both species. Notably, we identified MARCKS, CXCL12, PDGFRA, and LEPR together with adipogenic factors as archetypal biomarkers of native MSCs within BM. In addition, our data suggest some complex gene nodes regulating critical biological functions of native BM-MSCs together with a preferential commitment toward an adipocyte lineage. CONCLUSIONS Overall, our taxonomy for native BM non-hematopoietic compartment provides an explicit depiction of gene expression in human ECs, MCs and MSCs at single-cell resolution. This analysis helps enhance our understanding of the phenotype and the complexity of biological functions of native human BM-MSCs.
Collapse
Affiliation(s)
- Loïc Fiévet
- RESTORE, Université de Toulouse, EFS Occitanie, INP-ENVT, Inserm U1301, UMR CNRS 5070, France, Université de Toulouse, Toulouse, France
- CHU de Toulouse, IFB, Hôpital Purpan, Toulouse, France
| | - Nicolas Espagnolle
- RESTORE, Université de Toulouse, EFS Occitanie, INP-ENVT, Inserm U1301, UMR CNRS 5070, France, Université de Toulouse, Toulouse, France
| | - Daniela Gerovska
- Group of Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, 20014, San Sebastián, Spain
- Basque Foundation for Science, IKERBASQUE, 48009, Bilbao, Spain
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of Basque Country (UPV/EHU), 48940, Leioa, Spain
| | - David Bernard
- RESTORE, Université de Toulouse, EFS Occitanie, INP-ENVT, Inserm U1301, UMR CNRS 5070, France, Université de Toulouse, Toulouse, France
| | - Charlotte Syrykh
- Department d'Anatomie Pathologique, Institut Universitaire du Cancer, CHU de Toulouse, Toulouse, France
| | - Camille Laurent
- Department d'Anatomie Pathologique, Institut Universitaire du Cancer, CHU de Toulouse, Toulouse, France
| | - Pierre Layrolle
- Tonic Inserm/UPS UMR 1214, CHU Purpan Hospital, Toulouse, France
- UMR 1238 Inserm, Phy-OS, Bone Sarcoma and Remodeling of Calcified Tissues, School of Medicine, University of Nantes, Nantes, France
| | - Julien De Lima
- UMR 1238 Inserm, Phy-OS, Bone Sarcoma and Remodeling of Calcified Tissues, School of Medicine, University of Nantes, Nantes, France
| | - Arthur Justo
- Department de Chirurgie Orthopédique, Pierre Paul Riquet, Hôpital Purpan, Toulouse, France
| | - Nicolas Reina
- Department de Chirurgie Orthopédique, Pierre Paul Riquet, Hôpital Purpan, Toulouse, France
| | - Louis Casteilla
- RESTORE, Université de Toulouse, EFS Occitanie, INP-ENVT, Inserm U1301, UMR CNRS 5070, France, Université de Toulouse, Toulouse, France
| | - Marcos J Araúzo-Bravo
- Group of Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, 20014, San Sebastián, Spain
- Basque Foundation for Science, IKERBASQUE, 48009, Bilbao, Spain
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of Basque Country (UPV/EHU), 48940, Leioa, Spain
| | - Abderrahim Naji
- Department of Environmental Medicine, Cooperative Medicine Unit, Research and Education Faculty, Medicine Science Cluster, Nankoku, Kochi Prefecture, Japan
| | - Jean-Christophe Pagès
- RESTORE, Université de Toulouse, EFS Occitanie, INP-ENVT, Inserm U1301, UMR CNRS 5070, France, Université de Toulouse, Toulouse, France
- CHU de Toulouse, IFB, Hôpital Purpan, Toulouse, France
| | - Frédéric Deschaseaux
- RESTORE, Université de Toulouse, EFS Occitanie, INP-ENVT, Inserm U1301, UMR CNRS 5070, France, Université de Toulouse, Toulouse, France.
| |
Collapse
|
12
|
Zhao L, Lee AS, Sasagawa K, Sokol J, Wang Y, Ransom RC, Zhao X, Ma C, Steininger HM, Koepke LS, Borrelli MR, Brewer RE, Lee LL, Huang X, Ambrosi TH, Sinha R, Hoover MY, Seita J, Weissman IL, Wu JC, Wan DC, Xiao J, Longaker MT, Nguyen PK, Chan CK. A Combination of Distinct Vascular Stem/Progenitor Cells for Neovascularization and Ischemic Rescue. Arterioscler Thromb Vasc Biol 2023; 43:1262-1277. [PMID: 37051932 PMCID: PMC10281192 DOI: 10.1161/atvbaha.122.317943] [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: 12/10/2021] [Revised: 03/09/2023] [Accepted: 03/28/2023] [Indexed: 04/14/2023]
Abstract
BACKGROUND Peripheral vascular disease remains a leading cause of vascular morbidity and mortality worldwide despite advances in medical and surgical therapy. Besides traditional approaches, which can only restore blood flow to native arteries, an alternative approach is to enhance the growth of new vessels, thereby facilitating the physiological response to ischemia. METHODS The ActinCreER/R26VT2/GK3 Rainbow reporter mouse was used for unbiased in vivo survey of injury-responsive vasculogenic clonal formation. Prospective isolation and transplantation were used to determine vessel-forming capacity of different populations. Single-cell RNA-sequencing was used to characterize distinct vessel-forming populations and their interactions. RESULTS Two populations of distinct vascular stem/progenitor cells (VSPCs) were identified from adipose-derived mesenchymal stromal cells: VSPC1 is CD45-Ter119-Tie2+PDGFRa-CD31+CD105highSca1low, which gives rise to stunted vessels (incomplete tubular structures) in a transplant setting, and VSPC2 which is CD45-Ter119-Tie2+PDGFRa+CD31-CD105lowSca1high and forms stunted vessels and fat. Interestingly, cotransplantation of VSPC1 and VSPC2 is required to form functional vessels that improve perfusion in the mouse hindlimb ischemia model. Similarly, VSPC1 and VSPC2 populations isolated from human adipose tissue could rescue the ischemic condition in mice. CONCLUSIONS These findings suggest that autologous cotransplantation of synergistic VSPCs from nonessential adipose tissue can promote neovascularization and represents a promising treatment for ischemic disease.
Collapse
Affiliation(s)
- Liming Zhao
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (L.Z., Y.W., J.X.)
| | - Andrew S. Lee
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, China (A.S.L.)
- Institute for Cancer Research, Shenzhen Bay Laboratory, China (A.S.L.)
| | - Koki Sasagawa
- Stanford Cardiovascular Institute (K.S., J.S., X.Z., X.H., J.C.W., M.T.L., P.K.N., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Medicine, Division of Cardiovascular Medicine (K.S., J.S., X.Z., X.H., J.C.W., P.K.N.), Stanford University School of Medicine, CA
| | - Jan Sokol
- Stanford Cardiovascular Institute (K.S., J.S., X.Z., X.H., J.C.W., M.T.L., P.K.N., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Medicine, Division of Cardiovascular Medicine (K.S., J.S., X.Z., X.H., J.C.W., P.K.N.), Stanford University School of Medicine, CA
- Center for Integrative Medical Sciences and Advanced Data Science Project, RIKEN, Tokyo, Japan (J.S.)
| | - Yuting Wang
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (L.Z., Y.W., J.X.)
| | - Ryan C. Ransom
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Xin Zhao
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Stanford Cardiovascular Institute (K.S., J.S., X.Z., X.H., J.C.W., M.T.L., P.K.N., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Medicine, Division of Cardiovascular Medicine (K.S., J.S., X.Z., X.H., J.C.W., P.K.N.), Stanford University School of Medicine, CA
| | - Chao Ma
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Holly M. Steininger
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Lauren S. Koepke
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Mimi R. Borrelli
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Rachel E. Brewer
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Lorene L.Y. Lee
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Xianxi Huang
- Stanford Cardiovascular Institute (K.S., J.S., X.Z., X.H., J.C.W., M.T.L., P.K.N., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Medicine, Division of Cardiovascular Medicine (K.S., J.S., X.Z., X.H., J.C.W., P.K.N.), Stanford University School of Medicine, CA
| | - Thomas H. Ambrosi
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Rahul Sinha
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Malachia Y. Hoover
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Jun Seita
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Stanford Cardiovascular Institute (K.S., J.S., X.Z., X.H., J.C.W., M.T.L., P.K.N., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Medicine, Division of Cardiovascular Medicine (K.S., J.S., X.Z., X.H., J.C.W., P.K.N.), Stanford University School of Medicine, CA
- Department of Developmental Biology (I.L.W., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (L.Z., Y.W., J.X.)
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, China (A.S.L.)
- Institute for Cancer Research, Shenzhen Bay Laboratory, China (A.S.L.)
- Center for Integrative Medical Sciences and Advanced Data Science Project, RIKEN, Tokyo, Japan (J.S.)
| | - Irving L. Weissman
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Developmental Biology (I.L.W., C.K.F.C.), Stanford University School of Medicine, CA
| | - Joseph C. Wu
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Stanford Cardiovascular Institute (K.S., J.S., X.Z., X.H., J.C.W., M.T.L., P.K.N., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Medicine, Division of Cardiovascular Medicine (K.S., J.S., X.Z., X.H., J.C.W., P.K.N.), Stanford University School of Medicine, CA
| | - Derrick C. Wan
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Jun Xiao
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (L.Z., Y.W., J.X.)
| | - Michael T. Longaker
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
| | - Patricia K. Nguyen
- Stanford Cardiovascular Institute (K.S., J.S., X.Z., X.H., J.C.W., M.T.L., P.K.N., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Medicine, Division of Cardiovascular Medicine (K.S., J.S., X.Z., X.H., J.C.W., P.K.N.), Stanford University School of Medicine, CA
| | - Charles K.F. Chan
- Institute for Stem Cell Biology and Regenerative Medicine (L.Z., Y.W., R.C.R., X.Z., C.M., H.M.S., L.S.K., M.R.B., R.E.B., L.Y.L., T.H.A., R.S., M.Y.H., I.L.W., J.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Surgery, Division of Plastic and Reconstructive Surgery (L.Z., Y.W., R.C.R., C.M., H.M.S., L.S.K., M.R.B., L.L.Y.L., T.H.A., D.C.W., M.T.L., C.K.F.C.), Stanford University School of Medicine, CA
- Department of Developmental Biology (I.L.W., C.K.F.C.), Stanford University School of Medicine, CA
| |
Collapse
|
13
|
Shen F, Huang X, He G, Shi Y. The emerging studies on mesenchymal progenitors in the long bone. Cell Biosci 2023; 13:105. [PMID: 37301964 DOI: 10.1186/s13578-023-01039-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 05/01/2023] [Indexed: 06/12/2023] Open
Abstract
Mesenchymal progenitors (MPs) are considered to play vital roles in bone development, growth, bone turnover, and repair. In recent years, benefiting from advanced approaches such as single-cell sequence, lineage tracing, flow cytometry, and transplantation, multiple MPs are identified and characterized in several locations of bone, including perichondrium, growth plate, periosteum, endosteum, trabecular bone, and stromal compartment. However, although great discoveries about skeletal stem cells (SSCs) and progenitors are present, it is still largely obscure how the varied landscape of MPs from different residing sites diversely contribute to the further differentiation of osteoblasts, osteocytes, chondrocytes, and other stromal cells in their respective destiny sites during development and regeneration. Here we discuss recent findings on MPs' origin, differentiation, and maintenance during long bone development and homeostasis, providing clues and models of how the MPs contribute to bone development and repair.
Collapse
Affiliation(s)
- Fangyuan Shen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaobin Huang
- Department of Oral and Maxillofacial Surgery/Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guangxu He
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, NO. 139 Middle Renmin Road, Changsha, Hunan, China.
| | - Yu Shi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
14
|
Rong L, Zhang L, Yang Z, Xu L. New insights into the properties, functions, and aging of skeletal stem cells. Osteoporos Int 2023:10.1007/s00198-023-06736-4. [PMID: 37069243 DOI: 10.1007/s00198-023-06736-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 03/27/2023] [Indexed: 04/19/2023]
Abstract
Bone-related diseases pose a major health burden for modern society. Bone is one of the organs that rely on stem cell function to maintain tissue homeostasis. Stem cell therapy has emerged as an effective new strategy to repair and replace damaged tissue. Although research on bone marrow mesenchymal stem cells has been conducted over the last few decades, the identity and definition of the true skeletal stem cell population remains controversial. Due to technological advances, some progress has been made in the prospective separation and function research of purified skeletal stem cells. Here, we reviewed the recent progress of highly purified skeletal stem cells, their function in bone development and repair, and the impact of aging on skeletal stem cells. Various studies on animal and human models distinguished and isolated skeletal stem cells using different surface markers based on flow-cytometry-activated cell sorting. The roles of different types of skeletal stem cells in bone growth, remodeling, and repair are gradually becoming clear. Thanks to technological advances, SSCs can be specifically identified and purified for functional testing and molecular analysis. The basic features of SSCs and their roles in bone development and repair and the effects of aging on SSCs are gradually being elucidated. Future mechanistic studies can help to develop new therapeutic interventions to improve various types of skeletal diseases and enhance the regenerative potential of SSCs.
Collapse
Affiliation(s)
- Lingjun Rong
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lixia Zhang
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zaigang Yang
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Lijun Xu
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
15
|
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
|
16
|
Solidum JGN, Jeong Y, Heralde F, Park D. Differential regulation of skeletal stem/progenitor cells in distinct skeletal compartments. Front Physiol 2023; 14:1137063. [PMID: 36926193 PMCID: PMC10013690 DOI: 10.3389/fphys.2023.1137063] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 02/16/2023] [Indexed: 03/06/2023] Open
Abstract
Skeletal stem/progenitor cells (SSPCs), characterized by self-renewal and multipotency, are essential for skeletal development, bone remodeling, and bone repair. These cells have traditionally been known to reside within the bone marrow, but recent studies have identified the presence of distinct SSPC populations in other skeletal compartments such as the growth plate, periosteum, and calvarial sutures. Differences in the cellular and matrix environment of distinct SSPC populations are believed to regulate their stemness and to direct their roles at different stages of development, homeostasis, and regeneration; differences in embryonic origin and adjacent tissue structures also affect SSPC regulation. As these SSPC niches are dynamic and highly specialized, changes under stress conditions and with aging can alter the cellular composition and molecular mechanisms in place, contributing to the dysregulation of local SSPCs and their activity in bone regeneration. Therefore, a better understanding of the different regulatory mechanisms for the distinct SSPCs in each skeletal compartment, and in different conditions, could provide answers to the existing knowledge gap and the impetus for realizing their potential in this biological and medical space. Here, we summarize the current scientific advances made in the study of the differential regulation pathways for distinct SSPCs in different bone compartments. We also discuss the physical, biological, and molecular factors that affect each skeletal compartment niche. Lastly, we look into how aging influences the regenerative capacity of SSPCs. Understanding these regulatory differences can open new avenues for the discovery of novel treatment approaches for calvarial or long bone repair.
Collapse
Affiliation(s)
- Jea Giezl Niedo Solidum
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Manila, Philippines
- Department of Molecular and Human Genetics, Houston, TX, United States
| | - Youngjae Jeong
- Department of Molecular and Human Genetics, Houston, TX, United States
| | - Francisco Heralde
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Dongsu Park
- Department of Molecular and Human Genetics, Houston, TX, United States
- Center for Skeletal Biology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
17
|
Denervation during mandibular distraction osteogenesis results in impaired bone formation. Sci Rep 2023; 13:2097. [PMID: 36747028 PMCID: PMC9902545 DOI: 10.1038/s41598-023-27921-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 01/10/2023] [Indexed: 02/08/2023] Open
Abstract
Mandibular distraction osteogenesis (DO) is mediated by skeletal stem cells (SSCs) in mice, which enact bone regeneration via neural crest re-activation. As peripheral nerves are essential to progenitor function during development and in response to injury, we questioned if denervation impairs mandibular DO. C57Bl6 mice were divided into two groups: DO with a segmental defect in the inferior alveolar nerve (IAN) at the time of mandibular osteotomy ("DO Den") and DO with IAN intact ("DO Inn"). DO Den demonstrated significantly reduced histological and radiological osteogenesis relative to DO Inn. Denervation preceding DO results in reduced SSC amplification and osteogenic potential in mice. Single cell RNA sequencing analysis revealed that there was a predominance of innervated SSCs in clusters dominated by pathways related to bone formation. A rare human patient specimen was also analyzed and suggested that histological, radiological, and transcriptional alterations seen in mouse DO may be conserved in the setting of denervated human mandible distraction. Fibromodulin (FMOD) transcriptional and protein expression were reduced in denervated relative to innervated mouse and human mandible regenerate. Finally, when exogenous FMOD was added to DO-Den and DO-Inn SSCs undergoing in vitro osteogenic differentiation, the osteogenic potential of DO-Den SSCs was increased in comparison to control untreated DO-Den SSCs, modeling the superior osteogenic potential of DO-Inn SSCs.
Collapse
|
18
|
Andrew TW, Koepke LS, Wang Y, Lopez M, Steininger H, Struck D, Boyko T, Ambrosi TH, Tong X, Sun Y, Gulati GS, Murphy MP, Marecic O, Tevlin R, Schallmoser K, Strunk D, Seita J, Goodman SB, Yang F, Longaker MT, Yang GP, Chan CKF. Sexually dimorphic estrogen sensing in skeletal stem cells controls skeletal regeneration. Nat Commun 2022; 13:6491. [PMID: 36310174 PMCID: PMC9618571 DOI: 10.1038/s41467-022-34063-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 10/05/2022] [Indexed: 12/25/2022] Open
Abstract
Sexually dimorphic tissues are formed by cells that are regulated by sex hormones. While a number of systemic hormones and transcription factors are known to regulate proliferation and differentiation of osteoblasts and osteoclasts, the mechanisms that determine sexually dimorphic differences in bone regeneration are unclear. To explore how sex hormones regulate bone regeneration, we compared bone fracture repair between adult male and female mice. We found that skeletal stem cell (SSC) mediated regeneration in female mice is dependent on estrogen signaling but SSCs from male mice do not exhibit similar estrogen responsiveness. Mechanistically, we found that estrogen acts directly on the SSC lineage in mice and humans by up-regulating multiple skeletogenic pathways and is necessary for the stem cell's ability to self- renew and differentiate. Our results also suggest a clinically applicable strategy to accelerate bone healing using localized estrogen hormone therapy.
Collapse
Affiliation(s)
- Tom W Andrew
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Lauren S Koepke
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Yuting Wang
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Michael Lopez
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Holly Steininger
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Danielle Struck
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Tatiana Boyko
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Thomas H Ambrosi
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Xinming Tong
- Department of Bioengineering, Stanford University, Palo Alto, CA, 94305, USA
| | - Yuxi Sun
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
- Birmingham VA Medical Center, Birmingham, AL, 35233, USA
| | - Gunsagar S Gulati
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Matthew P Murphy
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Owen Marecic
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Ruth Tevlin
- Division of Plastic and Reconstructive Surgery, Stanford Hospital and Clinics, Palo Alto, CA, USA
| | - Katharina Schallmoser
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Department for Transfusion Medicine, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Dirk Strunk
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Department for Transfusion Medicine, Paracelsus Medical University of Salzburg, Salzburg, Austria
- Cell Therapy Institute, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Jun Seita
- Center for Integrative Medical Sciences and Advanced Data Science Project, RIKEN, Tokyo, Japan
| | - Stuart B Goodman
- Department of Orthopedic Surgery, Stanford University, Palo Alto, CA, 94305, USA
| | - Fan Yang
- Department of Bioengineering, Stanford University, Palo Alto, CA, 94305, USA
- Department of Orthopedic Surgery, Stanford University, Palo Alto, CA, 94305, USA
| | - Michael T Longaker
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - George P Yang
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
- Birmingham VA Medical Center, Birmingham, AL, 35233, USA.
| | - Charles K F Chan
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
19
|
Abstract
The tissue-resident skeletal stem cells (SSCs), which are self-renewal and multipotent, continuously provide cells (including chondrocytes, bone cells, marrow adipocytes, and stromal cells) for the development and homeostasis of the skeletal system. In recent decade, utilizing fluorescence-activated cell sorting, lineage tracing, and single-cell sequencing, studies have identified various types of SSCs, plotted the lineage commitment trajectory, and partially revealed their properties under physiological and pathological conditions. In this review, we retrospect to SSCs identification and functional studies. We discuss the principles and approaches to identify bona fide SSCs, highlighting pioneering findings that plot the lineage atlas of SSCs. The roles of SSCs and progenitors in long bone, craniofacial tissues, and periosteum are systematically discussed. We further focus on disputes and challenges in SSC research.
Collapse
|
20
|
Functional Heterogeneity of Bone Marrow Mesenchymal Stem Cell Subpopulations in Physiology and Pathology. Int J Mol Sci 2022; 23:ijms231911928. [PMID: 36233230 PMCID: PMC9570000 DOI: 10.3390/ijms231911928] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs) are multi-potent cell populations and are capable of maintaining bone and body homeostasis. The stemness and potential therapeutic effect of BMSCs have been explored extensively in recent years. However, diverse cell surface antigens and complex gene expression of BMSCs have indicated that BMSCs represent heterogeneous populations, and the natural characteristics of BMSCs make it difficult to identify the specific subpopulations in pathological processes which are often obscured by bulk analysis of the total BMSCs. Meanwhile, the therapeutic effect of total BMSCs is often less effective partly due to their heterogeneity. Therefore, understanding the functional heterogeneity of the BMSC subpopulations under different physiological and pathological conditions could have major ramifications for global health. Here, we summarize the recent progress of functional heterogeneity of BMSC subpopulations in physiology and pathology. Targeting tissue-resident single BMSC subpopulation offers a potentially innovative therapeutic strategy and improves BMSC effectiveness in clinical application.
Collapse
|
21
|
Kim MJ, Valderrábano RJ, Wu JY. Osteoblast Lineage Support of Hematopoiesis in Health and Disease. J Bone Miner Res 2022; 37:1823-1842. [PMID: 35983701 PMCID: PMC11346465 DOI: 10.1002/jbmr.4678] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/21/2022] [Accepted: 08/13/2022] [Indexed: 11/06/2022]
Abstract
In mammals, hematopoiesis migrates to the bone marrow during embryogenesis coincident with the appearance of mineralized bone, where hematopoietic stem cells (HSCs) and their progeny are maintained by the surrounding microenvironment or niche, and sustain the entirety of the hematopoietic system. Genetic manipulation of niche factors and advances in cell lineage tracing techniques have implicated cells of both hematopoietic and nonhematopoietic origin as important regulators of hematopoiesis in health and disease. Among them, cells of the osteoblast lineage, from stromal skeletal stem cells to matrix-embedded osteocytes, are vital niche residents with varying capacities for hematopoietic support depending on stage of differentiation. Here, we review populations of osteoblasts at differing stages of differentiation and summarize the current understanding of the role of the osteoblast lineage in supporting hematopoiesis. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Matthew J Kim
- Division of Endocrinology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Rodrigo J Valderrábano
- Research Program in Men's Health: Aging and Metabolism, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joy Y Wu
- Division of Endocrinology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
22
|
Ferreira MJS, Mancini FE, Humphreys PA, Ogene L, Buckley M, Domingos MAN, Kimber SJ. Pluripotent stem cells for skeletal tissue engineering. Crit Rev Biotechnol 2022; 42:774-793. [PMID: 34488516 DOI: 10.1080/07388551.2021.1968785] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Here, we review the use of human pluripotent stem cells for skeletal tissue engineering. A number of approaches have been used for generating cartilage and bone from both human embryonic stem cells and induced pluripotent stem cells. These range from protocols relying on intrinsic cell interactions and signals from co-cultured cells to those attempting to recapitulate the series of steps occurring during mammalian skeletal development. The importance of generating authentic tissues rather than just differentiated cells is emphasized and enabling technologies for doing this are reported. We also review the different methods for characterization of skeletal cells and constructs at the tissue and single-cell level, and indicate newer resources not yet fully utilized in this field. There have been many challenges in this research area but the technologies to overcome these are beginning to appear, often adopted from related fields. This makes it more likely that cost-effective and efficacious human pluripotent stem cell-engineered constructs may become available for skeletal repair in the near future.
Collapse
Affiliation(s)
- Miguel J S Ferreira
- Department of Mechanical, Aerospace and Civil Engineering, School of Engineering, Faculty of Science and Engineering & Henry Royce Institute, The University of Manchester, Manchester, UK
| | - Fabrizio E Mancini
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Paul A Humphreys
- Department of Mechanical, Aerospace and Civil Engineering, School of Engineering, Faculty of Science and Engineering & Henry Royce Institute, The University of Manchester, Manchester, UK
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Leona Ogene
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Michael Buckley
- Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | - Marco A N Domingos
- Department of Mechanical, Aerospace and Civil Engineering, School of Engineering, Faculty of Science and Engineering & Henry Royce Institute, The University of Manchester, Manchester, UK
| | - Susan J Kimber
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| |
Collapse
|
23
|
Titan AL, Davitt M, Foster D, Salhotra A, Menon S, Chen K, Fahy E, Lopez M, Jones RE, Baiu I, Burcham A, Januszyk M, Gurtner G, Fox P, Chan C, Quarto N, Longaker M. Partial Tendon Injury at the Tendon-to-Bone Enthesis Activates Skeletal Stem Cells. Stem Cells Transl Med 2022; 11:715-726. [PMID: 35640155 PMCID: PMC9299518 DOI: 10.1093/stcltm/szac027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 03/09/2022] [Indexed: 11/24/2022] Open
Abstract
The tendon enthesis plays a critical role in facilitating movement and reducing stress within joints. Partial enthesis injuries heal in a mechanically inferior manner and never achieve healthy tissue function. The cells responsible for tendon-to-bone healing remain incompletely characterized and their origin is unknown. Here, we evaluated the putative role of mouse skeletal stem cells (mSSCs) in the enthesis after partial-injury. We found that mSSCs were present at elevated levels within the enthesis following injury and that these cells downregulated TGFβ signaling pathway elements at both the RNA and protein levels. Exogenous application of TGFβ post-injury led to a reduced mSSC response and impaired healing, whereas treatment with a TGFβ inhibitor (SB43154) resulted in a more robust mSSC response. Collectively, these data suggest that mSSCs may augment tendon-to-bone healing by dampening the effects of TGFβ signaling within the mSSC niche.
Collapse
Affiliation(s)
- Ashley L Titan
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Davitt
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Deshka Foster
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Ankit Salhotra
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Siddharth Menon
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Kellen Chen
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Evan Fahy
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Lopez
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - R Ellen Jones
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Ioana Baiu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Austin Burcham
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Januszyk
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Geoffrey Gurtner
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Paige Fox
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Charles Chan
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Natalina Quarto
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA
| |
Collapse
|
24
|
Soliman H, Theret M, Scott W, Hill L, Underhill TM, Hinz B, Rossi FMV. Multipotent stromal cells: One name, multiple identities. Cell Stem Cell 2021; 28:1690-1707. [PMID: 34624231 DOI: 10.1016/j.stem.2021.09.001] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Multipotent stromal cells (MSCs) are vital for development, maintenance, function, and regeneration of most tissues. They can differentiate along multiple connective lineages, but unlike most other stem/progenitor cells, they carry out various other functions while maintaining their developmental potential. MSCs function as damage sensors, respond to injury by fostering regeneration through secretion of trophic factors as well as extracellular matrix (ECM) molecules, and contribute to fibrotic reparative processes when regeneration fails. Tissue-specific MSC identity, fate(s), and function(s) are being resolved through fate mapping coupled with single cell "omics," providing unparalleled insights into the secret lives of tissue-resident MSCs.
Collapse
Affiliation(s)
- Hesham Soliman
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Aspect Biosystems, Vancouver, BC V6P 6P2, Canada
| | - Marine Theret
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Wilder Scott
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Lesley Hill
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Tully Michael Underhill
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Fabio M V Rossi
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
25
|
Ambrosi TH, Marecic O, McArdle A, Sinha R, Gulati GS, Tong X, Wang Y, Steininger HM, Hoover MY, Koepke LS, Murphy MP, Sokol J, Seo EY, Tevlin R, Lopez M, Brewer RE, Mascharak S, Lu L, Ajanaku O, Conley SD, Seita J, Morri M, Neff NF, Sahoo D, Yang F, Weissman IL, Longaker MT, Chan CKF. Aged skeletal stem cells generate an inflammatory degenerative niche. Nature 2021; 597:256-262. [PMID: 34381212 PMCID: PMC8721524 DOI: 10.1038/s41586-021-03795-7] [Citation(s) in RCA: 210] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/05/2021] [Indexed: 12/22/2022]
Abstract
Loss of skeletal integrity during ageing and disease is associated with an imbalance in the opposing actions of osteoblasts and osteoclasts1. Here we show that intrinsic ageing of skeletal stem cells (SSCs)2 in mice alters signalling in the bone marrow niche and skews the differentiation of bone and blood lineages, leading to fragile bones that regenerate poorly. Functionally, aged SSCs have a decreased bone- and cartilage-forming potential but produce more stromal lineages that express high levels of pro-inflammatory and pro-resorptive cytokines. Single-cell RNA-sequencing studies link the functional loss to a diminished transcriptomic diversity of SSCs in aged mice, which thereby contributes to the transformation of the bone marrow niche. Exposure to a youthful circulation through heterochronic parabiosis or systemic reconstitution with young haematopoietic stem cells did not reverse the diminished osteochondrogenic activity of aged SSCs, or improve bone mass or skeletal healing parameters in aged mice. Conversely, the aged SSC lineage promoted osteoclastic activity and myeloid skewing by haematopoietic stem and progenitor cells, suggesting that the ageing of SSCs is a driver of haematopoietic ageing. Deficient bone regeneration in aged mice could only be returned to youthful levels by applying a combinatorial treatment of BMP2 and a CSF1 antagonist locally to fractures, which reactivated aged SSCs and simultaneously ablated the inflammatory, pro-osteoclastic milieu. Our findings provide mechanistic insights into the complex, multifactorial mechanisms that underlie skeletal ageing and offer prospects for rejuvenating the aged skeletal system.
Collapse
Affiliation(s)
- Thomas H Ambrosi
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Owen Marecic
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Adrian McArdle
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Rahul Sinha
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Gunsagar S Gulati
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Xinming Tong
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Yuting Wang
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Holly M Steininger
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Malachia Y Hoover
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Lauren S Koepke
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Matthew P Murphy
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Jan Sokol
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Eun Young Seo
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Ruth Tevlin
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Lopez
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Rachel E Brewer
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Shamik Mascharak
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Laura Lu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Oyinkansola Ajanaku
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Stephanie D Conley
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jun Seita
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Center for Integrative Medical Sciences and Advanced Data Science Project, RIKEN, Tokyo, Japan
| | | | | | - Debashis Sahoo
- Pediatrics, and Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA
| | - Fan Yang
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Irving L Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Ludwig Center for Cancer Stem Cell Biology and Medicine at Stanford University, Stanford, CA, USA
| | - Michael T Longaker
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| | - Charles K F Chan
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
26
|
Yamaguchi A, Hirano I, Narusawa S, Shimizu K, Ariyama H, Yamawaki K, Nagao K, Yamamoto M, Shimizu R. Blockade of the interaction between BMP9 and endoglin on erythroid progenitors promotes erythropoiesis in mice. Genes Cells 2021; 26:782-797. [PMID: 34333851 PMCID: PMC9290798 DOI: 10.1111/gtc.12887] [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: 03/22/2021] [Revised: 07/26/2021] [Accepted: 07/30/2021] [Indexed: 01/19/2023]
Abstract
Bone morphogenetic protein‐9 (BMP9), a member of the transforming growth factor β (TGFβ) superfamily, plays important roles in the development and maintenance of various cell lineages via complexes of type I and type II TGFβ receptors. Endoglin is a coreceptor for several TGFβ family members, including BMP9, which is highly expressed in a particular stage of differentiation in erythroid cells as well as in endothelial cells. Although the importance of the interaction between BMP9 and endoglin for endothelial development has been reported, the contribution of BMP9 to endoglin‐expressing erythroid cells remains to be clarified. To address this point, we prepared an anti‐BMP9 antibody that blocks the BMP9‐endoglin interaction. Of note, challenge with the antibody promotes erythropoiesis in wild‐type mice but not in a mouse model of renal anemia in which erythropoietin (EPO) production in the kidneys is genetically ablated. While endoglin‐positive erythroid progenitors are mainly maintained as progenitors when bone marrow‐derived lineage‐negative and cKit‐positive cells are cultured in the presence of EPO and stem cell factor, the erythroid‐biased accumulation of progenitors is impeded by the presence of BMP9. Our findings uncover an unrecognized role for BMP9 in attenuating erythroid differentiation via its interaction with endoglin on erythroid progenitors.
Collapse
Affiliation(s)
- Ayami Yamaguchi
- Nephrology Research Labs., Nephrology R&D Unit, R&D Division, Kyowa Kirin Co., Ltd., Machida, Japan
| | - Ikuo Hirano
- Department of Molecular Hematology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shiho Narusawa
- Department of Molecular Hematology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kiyoshi Shimizu
- Nephrology Research Labs., Nephrology R&D Unit, R&D Division, Kyowa Kirin Co., Ltd., Machida, Japan
| | - Hiroyuki Ariyama
- Nephrology Research Labs., Nephrology R&D Unit, R&D Division, Kyowa Kirin Co., Ltd., Machida, Japan
| | - Kengo Yamawaki
- Nephrology Research Labs., Nephrology R&D Unit, R&D Division, Kyowa Kirin Co., Ltd., Machida, Japan
| | - Kenji Nagao
- Nephrology Research Labs., Nephrology R&D Unit, R&D Division, Kyowa Kirin Co., Ltd., Machida, Japan
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan.,Tohoku Medical Mega-Bank Organization, Tohoku University, Sendai, Japan
| | - Ritsuko Shimizu
- Department of Molecular Hematology, Tohoku University Graduate School of Medicine, Sendai, Japan.,Tohoku Medical Mega-Bank Organization, Tohoku University, Sendai, Japan
| |
Collapse
|
27
|
Cranial Suture Mesenchymal Stem Cells: Insights and Advances. Biomolecules 2021; 11:biom11081129. [PMID: 34439795 PMCID: PMC8392244 DOI: 10.3390/biom11081129] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/25/2021] [Accepted: 07/27/2021] [Indexed: 02/05/2023] Open
Abstract
The cranial bones constitute the protective structures of the skull, which surround and protect the brain. Due to the limited repair capacity, the reconstruction and regeneration of skull defects are considered as an unmet clinical need and challenge. Previously, it has been proposed that the periosteum and dura mater provide reparative progenitors for cranial bones homeostasis and injury repair. In addition, it has also been speculated that the cranial mesenchymal stem cells reside in the perivascular niche of the diploe, namely, the soft spongy cancellous bone between the interior and exterior layers of cortical bone of the skull, which resembles the skeletal stem cells’ distribution pattern of the long bone within the bone marrow. Not until recent years have several studies unraveled and validated that the major mesenchymal stem cell population of the cranial region is primarily located within the suture mesenchyme of the skull, and hence, they are termed suture mesenchymal stem cells (SuSCs). Here, we summarized the characteristics of SuSCs, this newly discovered stem cell population of cranial bones, including the temporospatial distribution pattern, self-renewal, and multipotent properties, contribution to injury repair, as well as the signaling pathways and molecular mechanisms associated with the regulation of SuSCs.
Collapse
|
28
|
Ambrosi TH, Sinha R, Steininger HM, Hoover MY, Murphy MP, Koepke LS, Wang Y, Lu WJ, Morri M, Neff NF, Weissman IL, Longaker MT, Chan CKF. Distinct skeletal stem cell types orchestrate long bone skeletogenesis. eLife 2021; 10:e66063. [PMID: 34280086 PMCID: PMC8289409 DOI: 10.7554/elife.66063] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 07/02/2021] [Indexed: 12/30/2022] Open
Abstract
Skeletal stem and progenitor cell populations are crucial for bone physiology. Characterization of these cell types remains restricted to heterogenous bulk populations with limited information on whether they are unique or overlap with previously characterized cell types. Here we show, through comprehensive functional and single-cell transcriptomic analyses, that postnatal long bones of mice contain at least two types of bone progenitors with bona fide skeletal stem cell (SSC) characteristics. An early osteochondral SSC (ocSSC) facilitates long bone growth and repair, while a second type, a perivascular SSC (pvSSC), co-emerges with long bone marrow and contributes to shape the hematopoietic stem cell niche and regenerative demand. We establish that pvSSCs, but not ocSSCs, are the origin of bone marrow adipose tissue. Lastly, we also provide insight into residual SSC heterogeneity as well as potential crosstalk between the two spatially distinct cell populations. These findings comprehensively address previously unappreciated shortcomings of SSC research.
Collapse
Affiliation(s)
- Thomas H Ambrosi
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| | - Rahul Sinha
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| | - Holly M Steininger
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| | - Malachia Y Hoover
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| | - Matthew P Murphy
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| | - Lauren S Koepke
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| | - Yuting Wang
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| | - Wan-Jin Lu
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| | | | - Norma F Neff
- Chan Zuckerberg BioHubSan FranciscoUnited States
| | - Irving L Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
- Ludwig Center for Cancer Stem Cell Biology and Medicine at Stanford UniversityStanfordUnited States
| | - Michael T Longaker
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
- Department of Surgery, Stanford University School of MedicineStanfordUnited States
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford UniversityStanfordUnited States
| | - Charles KF Chan
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
- Department of Surgery, Stanford University School of MedicineStanfordUnited States
| |
Collapse
|
29
|
Agarwal P, Li H, Choi K, Hueneman K, He J, Welner RS, Starczynowski DT, Bhatia R. TNF-α-induced alterations in stromal progenitors enhance leukemic stem cell growth via CXCR2 signaling. Cell Rep 2021; 36:109386. [PMID: 34260914 PMCID: PMC8292106 DOI: 10.1016/j.celrep.2021.109386] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/30/2021] [Accepted: 06/21/2021] [Indexed: 11/30/2022] Open
Abstract
Chronic myeloid leukemia (CML) is propagated by leukemia stem cells (LSCs) that are not eradicated by tyrosine kinase inhibitor (TKI) treatment and persist as a source of disease recurrence. Bone marrow (BM) mesenchymal niches play an essential role in hematopoietic stem cell (HSC) and LSC maintenance. Using a murine CML model, we examine leukemia-induced alterations in mesenchymal cell populations. We show that 6C3+ stromal progenitors expand in CML BM and exhibit increased LSC but reduced HSC supportive capacity. Tumor necrosis factor alpha (TNF-α) signaling mediates expansion and higher expression of CXCL1 in CML BM 6C3+ cells and higher expression of the CXCL1 receptor CXCR2 in LSCs. CXCL1 enhances LSC proliferation and self-renewal, whereas CXCR2 inhibition reduces LSC growth and enhances LSC targeting in combination with tyrosine kinase inhibitors (TKIs). We find that TNF-α-mediated alterations in CML BM stromal niches enhance support of LSC maintenance and growth via CXCL1-CXCR2 signaling and that CXCR2 inhibition effectively depletes CML LSCs.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Bone Marrow/pathology
- Cell Cycle/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Chemokines/metabolism
- Gene Expression Regulation, Leukemic/drug effects
- Hematopoietic Stem Cells/drug effects
- Hematopoietic Stem Cells/metabolism
- Humans
- Inflammation/genetics
- Inflammation/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Mesenchymal Stem Cells/drug effects
- Mesenchymal Stem Cells/metabolism
- Mice, Inbred C57BL
- Middle Aged
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Protein Kinase Inhibitors/pharmacology
- Receptors, Interleukin-8B/metabolism
- Signal Transduction/drug effects
- Tumor Necrosis Factor-alpha/metabolism
- Mice
Collapse
Affiliation(s)
- Puneet Agarwal
- Division of Hematology & Oncology, University of Alabama Birmingham, Birmingham, AL, USA; Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Hui Li
- Division of Hematology & Oncology, University of Alabama Birmingham, Birmingham, AL, USA
| | - Kwangmin Choi
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kathleen Hueneman
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jianbo He
- Division of Hematology & Oncology, University of Alabama Birmingham, Birmingham, AL, USA
| | - Robert S Welner
- Division of Hematology & Oncology, University of Alabama Birmingham, Birmingham, AL, USA
| | - Daniel T Starczynowski
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ravi Bhatia
- Division of Hematology & Oncology, University of Alabama Birmingham, Birmingham, AL, USA.
| |
Collapse
|
30
|
The characterization of distinct populations of murine skeletal cells that have different roles in B lymphopoiesis. Blood 2021; 138:304-317. [PMID: 33786586 DOI: 10.1182/blood.2020005865] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 03/20/2021] [Indexed: 02/06/2023] Open
Abstract
Hematopoiesis is extrinsically controlled by cells of the bone marrow microenvironment, including skeletal lineage cells. The identification and subsequent studies of distinct subpopulations of maturing skeletal cells is currently limited due to a lack of methods to isolate these cells. We found that murine Lineage-CD31-Sca-1-CD51+ cells can be divided into four subpopulations using flow cytometry, based on their expression of the platelet derived growth factor receptors ⍺ and β (PDGFR⍺ and PDGFRβ). The use of different skeletal lineage reporters confirmed the skeletal origin of the four populations. Multiplex immunohistochemistry studies revealed that all four populations were localized near the growth plate and trabecular bone and were rarely found near cortical bone regions or in central bone marrow. Functional studies revealed differences in their abundance, colony-forming unit-fibroblast capacity and potential to differentiate into mineralized osteoblasts or adipocytes in vitro. Furthermore, the four populations had distinct gene expression profiles and differential cell surface expression of leptin receptor (LEPR) and vascular cell adhesion molecule 1 (VCAM-1). Interestingly, we discovered that one of these four different skeletal populations showed the highest expression of genes involved in the extrinsic regulation of B lymphopoiesis. This cell population varied in abundance between distinct hematopoietically active skeletal sites, and significant differences in the proportions of B lymphocyte precursors were also observed in these distinct skeletal sites. It also supported pre-B lymphopoiesis in culture. Our method to isolate four distinct maturing skeletal populations will assist in elucidating the roles of distinct skeletal niche cells in regulating hematopoiesis and bone.
Collapse
|
31
|
Yahara Y, Ma X, Gracia L, Alman BA. Monocyte/Macrophage Lineage Cells From Fetal Erythromyeloid Progenitors Orchestrate Bone Remodeling and Repair. Front Cell Dev Biol 2021; 9:622035. [PMID: 33614650 PMCID: PMC7889961 DOI: 10.3389/fcell.2021.622035] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/12/2021] [Indexed: 12/21/2022] Open
Abstract
A third of the population sustains a bone fracture, and the pace of fracture healing slows with age. The slower pace of repair is responsible for the increased morbidity in older individuals who sustain a fracture. Bone healing progresses through overlapping phases, initiated by cells of the monocyte/macrophage lineage. The repair process ends with remodeling. This last phase is controlled by osteoclasts, which are bone-specific multinucleated cells also of the monocyte/macrophage lineage. The slower rate of healing in aging can be rejuvenated by macrophages from young animals, and secreted proteins from macrophage regulate undifferentiated mesenchymal cells to become bone-forming osteoblasts. Macrophages can derive from fetal erythromyeloid progenitors or from adult hematopoietic progenitors. Recent studies show that fetal erythromyeloid progenitors are responsible for the osteoclasts that form the space in bone for hematopoiesis and the fetal osteoclast precursors reside in the spleen postnatally, traveling through the blood to participate in fracture repair. Differences in secreted proteins between macrophages from old and young animals regulate the efficiency of osteoblast differentiation from undifferentiated mesenchymal precursor cells. Interestingly, during the remodeling phase osteoclasts can form from the fusion between monocyte/macrophage lineage cells from the fetal and postnatal precursor populations. Data from single cell RNA sequencing identifies specific markers for populations derived from the different precursor populations, a finding that can be used in future studies. Here, we review the diversity of macrophages and osteoclasts, and discuss recent finding about their developmental origin and functions, which provides novel insights into their roles in bone homeostasis and repair.
Collapse
Affiliation(s)
- Yasuhito Yahara
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, United States.,Department of Orthopaedic Surgery, Faculty of Medicine, University of Toyama, Toyama, Japan.,Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Xinyi Ma
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, United States.,Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| | - Liam Gracia
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, United States.,Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| | - Benjamin A Alman
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, United States.,Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
32
|
Agas D, Sabbieti MG. Archetypal autophagic players through new lenses for bone marrow stem/mature cells regulation. J Cell Physiol 2021; 236:6101-6114. [PMID: 33492700 DOI: 10.1002/jcp.30296] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/04/2021] [Accepted: 01/11/2021] [Indexed: 12/11/2022]
Abstract
The bone marrow landscape consists of specialized and stem/progenitor cells, which coordinate important tissue-related and systemic physiological features. Within the marrow cavity, stem/progenitor and differentiated hematopoietic and skeletal cells congregate into dynamic functional assemblies throughout specific anatomical regions, termed niches. There is a need for better understanding of the bone marrow microareas, through exploration of the intramural physical and molecular interactions of the distinctive cell populations. The elective liaisons established among the mesenchymal/stromal stem cell and hematopoietic stem cell lineage trees play a key role in orchestrating the stem/mature cell behavior and customized hierarchies within bone marrow cell populations. Recently, the autophagic apparatus has been discovered to be an important feature of bone marrow homeostasis. Autophagy-related factors involved in the labyrinthic and highly dynamic bone marrow workshop redesign the niche framework by coordinating the operational schedule of pluripotent stem and mature cells. The following report summarizes the most recent breakthroughs in our understanding of the intramural relationships between bone marrow cells and key autophagic mediators. Doubtless, the consideration of the autophagy-related and unrelated functions of main players, such as p62, Atg7, Atg5, and Beclin-1 remains a compelling task to thoroughly understand the complex relations between the heterogenic cell types that populate bone marrow.
Collapse
Affiliation(s)
- Dimitrios Agas
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Macerata, Italy
| | - Maria Giovanna Sabbieti
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Macerata, Italy
| |
Collapse
|
33
|
Ambrosi TH, Chan CKF. Skeletal Stem Cells as the Developmental Origin of Cellular Niches for Hematopoietic Stem and Progenitor Cells. Curr Top Microbiol Immunol 2021; 434:1-31. [PMID: 34850280 PMCID: PMC8864730 DOI: 10.1007/978-3-030-86016-5_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The skeletal system is a highly complex network of mesenchymal, hematopoietic, and vasculogenic stem cell lineages that coordinate the development and maintenance of defined microenvironments, so-called niches. Technological advancements in recent years have allowed for the dissection of crucial cell types as well as their autocrine and paracrine signals that regulate these niches during development, homeostasis, regeneration, and disease. Ingress of blood vessels and bone marrow hematopoiesis are initiated by skeletal stem cells (SSCs) and their more committed downstream lineage cell types that direct shape and form of skeletal elements. In this chapter, we focus on the role of SSCs as the developmental origin of niches for hematopoietic stem and progenitor cells. We discuss latest updates in the definition of SSCs, cellular processes establishing and maintaining niches, as well as alterations of stem cell microenvironments promoting malignancies. We conclude with an outlook on future studies that could take advantage of SSC-niche engineering as a basis for the development of new therapeutic tools to not only treat bone-related diseases but also maladies stemming from derailed niche dynamics altering hematopoietic output.
Collapse
Affiliation(s)
- Thomas H Ambrosi
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Charles K F Chan
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
34
|
Hao Z, Li J, Li B, Alder KD, Cahill SV, Munger AM, Lee I, Kwon HK, Back J, Xu S, Kang MJ, Lee FY. Smoking Alters Inflammation and Skeletal Stem and Progenitor Cell Activity During Fracture Healing in Different Murine Strains. J Bone Miner Res 2021; 36:186-198. [PMID: 32866293 PMCID: PMC9057220 DOI: 10.1002/jbmr.4175] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/16/2020] [Accepted: 08/27/2020] [Indexed: 12/14/2022]
Abstract
Smokers are at a higher risk of delayed union or nonunion after fracture repair. Few specific interventions are available for prevention because the molecular mechanisms that result in these negative sequelae are poorly understood. Murine models that mimic fracture healing in smokers are crucial in further understanding the local cellular and molecular alterations during fracture healing caused by smoking. We exposed three murine strains, C57BL/6J, 129X1/SvJ, and BALB/cJ, to cigarette smoke for 3 months before the induction of a midshaft transverse femoral osteotomy. We evaluated fracture healing 4 weeks after the osteotomy using radiography, micro-computed tomography (μCT), and biomechanical testing. Radiographic analysis demonstrated a significant decrease in the fracture healing capacity of smoking 129X1/SvJ mice. μCT results showed delayed remodeling of fracture calluses in all three strains after cigarette smoke exposure. Biomechanical testing indicated the most significant impairment in the functional properties of 129X1/SvJ in comparison with C57BL/6J and BALB/cJ mice after cigarette smoke exposure. Thus, the 129X1/SvJ strain is most suitable in simulating smoking-induced impaired fracture healing. Furthermore, in smoking 129X1/SvJ murine models, we investigated the molecular and cellular alterations in fracture healing caused by cigarette smoking using histology, flow cytometry, and multiplex cytokine/chemokine analysis. Histological analysis showed impaired chondrogenesis in cigarette smoking. In addition, the important reparative cell populations, including skeletal stem cells and their downstream progenitors, demonstrated decreased expansion after injury as a result of cigarette smoking. Moreover, significantly increased pro-inflammatory mediators and the recruitment of immune cells in fracture hematomas were demonstrated in smoking mice. Collectively, our findings demonstrate the significant cellular and molecular alterations during fracture healing impaired by smoking, including disrupted chondrogenesis, aberrant skeletal stem and progenitor cell activity, and a pronounced initial inflammatory response. © 2020 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Zichen Hao
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
- Department of Emergency and Trauma, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Jun Li
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
- Department of Orthopaedics, The Second Hospital of Anhui Medical University, Hefei, China
| | - Bo Li
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
- Department of Orthopaedics, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Kareme D Alder
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
| | - Sean V Cahill
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
| | - Alana M Munger
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
| | - Inkyu Lee
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
- Department of Life Science, Chung-Ang University, Seoul, Republic of Korea
| | - Hyuk-Kwon Kwon
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
| | - JungHo Back
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
| | - Shuogui Xu
- Department of Emergency and Trauma, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Min-Jong Kang
- Department of Medicine, Pulmonary, Critical Care, and Sleep Medicine, Yale University, School of Medicine, New Haven, CT, USA
| | - Francis Y Lee
- Department of Orthopaedics and Rehabilitation, Yale University, School of Medicine, New Haven, CT, USA
| |
Collapse
|
35
|
Molecular Modulation of Fetal Liver Hematopoietic Stem Cell Mobilization into Fetal Bone Marrow in Mice. Stem Cells Int 2020; 2020:8885154. [PMID: 33381191 PMCID: PMC7755487 DOI: 10.1155/2020/8885154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 11/03/2020] [Accepted: 12/04/2020] [Indexed: 11/24/2022] Open
Abstract
Development of hematopoietic stem cells is a complex process, which has been extensively investigated. Hematopoietic stem cells (HSCs) in mouse fetal liver are highly expanded to prepare for mobilization of HSCs into the fetal bone marrow. It is not completely known how the fetal liver niche regulates HSC expansion without loss of self-renewal ability. We reviewed current progress about the effects of fetal liver niche, chemokine, cytokine, and signaling pathways on HSC self-renewal, proliferation, and expansion. We discussed the molecular regulations of fetal HSC expansion in mouse and zebrafish. It is also unknown how HSCs from the fetal liver mobilize, circulate, and reside into the fetal bone marrow niche. We reviewed how extrinsic and intrinsic factors regulate mobilization of fetal liver HSCs into the fetal bone marrow, which provides tools to improve HSC engraftment efficiency during HSC transplantation. Understanding the regulation of fetal liver HSC mobilization into the fetal bone marrow will help us to design proper clinical therapeutic protocol for disease treatment like leukemia during pregnancy. We prospect that fetal cells, including hepatocytes and endothelial and hematopoietic cells, might regulate fetal liver HSC expansion. Components from vascular endothelial cells and bones might also modulate the lodging of fetal liver HSCs into the bone marrow. The current review holds great potential to deeply understand the molecular regulations of HSCs in the fetal liver and bone marrow in mammals, which will be helpful to efficiently expand HSCs in vitro.
Collapse
|
36
|
Endoglin: An 'Accessory' Receptor Regulating Blood Cell Development and Inflammation. Int J Mol Sci 2020; 21:ijms21239247. [PMID: 33287465 PMCID: PMC7729465 DOI: 10.3390/ijms21239247] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 12/13/2022] Open
Abstract
Transforming growth factor-β1 (TGF-β1) is a pleiotropic factor sensed by most cells. It regulates a broad spectrum of cellular responses including hematopoiesis. In order to process TGF-β1-responses in time and space in an appropriate manner, there is a tight regulation of its signaling at diverse steps. The downstream signaling is mediated by type I and type II receptors and modulated by the ‘accessory’ receptor Endoglin also termed cluster of differentiation 105 (CD105). Endoglin was initially identified on pre-B leukemia cells but has received most attention due to its high expression on activated endothelial cells. In turn, Endoglin has been figured out as the causative factor for diseases associated with vascular dysfunction like hereditary hemorrhagic telangiectasia-1 (HHT-1), pre-eclampsia, and intrauterine growth restriction (IUPR). Because HHT patients often show signs of inflammation at vascular lesions, and loss of Endoglin in the myeloid lineage leads to spontaneous inflammation, it is speculated that Endoglin impacts inflammatory processes. In line, Endoglin is expressed on progenitor/precursor cells during hematopoiesis as well as on mature, differentiated cells of the innate and adaptive immune system. However, so far only pro-monocytes and macrophages have been in the focus of research, although Endoglin has been identified in many other immune system cell subsets. These findings imply a functional role of Endoglin in the maturation and function of immune cells. Aside the functional relevance of Endoglin in endothelial cells, CD105 is differentially expressed during hematopoiesis, arguing for a role of this receptor in the development of individual cell lineages. In addition, Endoglin expression is present on mature immune cells of the innate (i.e., macrophages and mast cells) and the adaptive (i.e., T-cells) immune system, further suggesting Endoglin as a factor that shapes immune responses. In this review, we summarize current knowledge on Endoglin expression and function in hematopoietic precursors and mature hematopoietic cells of different lineages.
Collapse
|
37
|
Cao Y, Buckels EJ, Matthews BG. Markers for Identification of Postnatal Skeletal Stem Cells In Vivo. Curr Osteoporos Rep 2020; 18:655-665. [PMID: 33034805 DOI: 10.1007/s11914-020-00622-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/29/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW The adult skeleton contains stem cells involved in growth, homeostasis, and healing. Mesenchymal or skeletal stem cells are proposed to provide precursors to osteoblasts, chondrocytes, marrow adipocytes, and stromal cells. We review the evidence for existence and functionality of different skeletal stem cell pools, and the tools available for identifying or targeting these populations in mouse and human tissues. RECENT FINDINGS Lineage tracing and single cell-based techniques in mouse models indicate that multiple pools of stem cells exist in postnatal bone. These include growth plate stem cells, stem and progenitor cells in the diaphysis, reticular cells that only form bone in response to injury, and injury-responsive periosteal stem cells. New staining protocols have also been described for prospective isolation of human skeletal stem cells. Several populations of postnatal skeletal stem and progenitor cells have been identified in mice, and we have an increasing array of tools to target these cells. Most Cre models lack a high degree of specificity to define single populations. Human studies are less advanced and require further efforts to refine methods for identifying stem and progenitor cells in adult bone.
Collapse
Affiliation(s)
- Ye Cao
- Department of Molecular Medicine and Pathology, University of Auckland, Private Bag 92-019, Auckland, 1142, New Zealand
| | - Emma J Buckels
- Department of Molecular Medicine and Pathology, University of Auckland, Private Bag 92-019, Auckland, 1142, New Zealand
| | - Brya G Matthews
- Department of Molecular Medicine and Pathology, University of Auckland, Private Bag 92-019, Auckland, 1142, New Zealand.
| |
Collapse
|
38
|
Tevlin R, Longaker MT, Wan DC. Skeletal Stem Cells-A Paradigm Shift in the Field of Craniofacial Bone Tissue Engineering. FRONTIERS IN DENTAL MEDICINE 2020; 1:596706. [PMID: 35664558 PMCID: PMC9161996 DOI: 10.3389/fdmed.2020.596706] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Defects of the craniofacial skeleton arise as a direct result of trauma, diseases, oncological resection, or congenital anomalies. Current treatment options are limited, highlighting the importance for developing new strategies to restore form, function, and aesthetics of missing or damaged bone in the face and the cranium. For optimal reconstruction, the goal is to replace "like with like." With the inherent challenges of existing options, there is a clear need to develop alternative strategies to reconstruct the craniofacial skeleton. The success of mesenchymal stem cell-based approaches has been hampered by high heterogeneity of transplanted cell populations with inconsistent preclinical and clinical trial outcomes. Here, we discuss the novel characterization and isolation of mouse skeletal stem cell (SSC) populations and their response to injury, systemic disease, and how their re-activation in vivo can contribute to tissue regeneration. These studies led to the characterization of human SSCs which are able to self-renew, give rise to increasingly fate restricted progenitors, and differentiate into bone, cartilage, and bone marrow stroma, all on the clonal level in vivo without prior in vitro culture. SSCs hold great potential for implementation in craniofacial bone tissue engineering and regenerative medicine. As we begin to better understand the diversity and the nature of skeletal stem and progenitor cells, there is a tangible future whereby a subset of human adult SSCs can be readily purified from bone or activated in situ with broad potential applications in craniofacial tissue engineering.
Collapse
Affiliation(s)
- Ruth Tevlin
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Michael T. Longaker
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Derrick C. Wan
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
39
|
Salhotra A, Shah HN, Levi B, Longaker MT. Mechanisms of bone development and repair. Nat Rev Mol Cell Biol 2020; 21:696-711. [PMID: 32901139 DOI: 10.1038/s41580-020-00279-w] [Citation(s) in RCA: 612] [Impact Index Per Article: 122.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2020] [Indexed: 12/19/2022]
Abstract
Bone development occurs through a series of synchronous events that result in the formation of the body scaffold. The repair potential of bone and its surrounding microenvironment - including inflammatory, endothelial and Schwann cells - persists throughout adulthood, enabling restoration of tissue to its homeostatic functional state. The isolation of a single skeletal stem cell population through cell surface markers and the development of single-cell technologies are enabling precise elucidation of cellular activity and fate during bone repair by providing key insights into the mechanisms that maintain and regenerate bone during homeostasis and repair. Increased understanding of bone development, as well as normal and aberrant bone repair, has important therapeutic implications for the treatment of bone disease and ageing-related degeneration.
Collapse
Affiliation(s)
- Ankit Salhotra
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Harsh N Shah
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Benjamin Levi
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
| | - Michael T Longaker
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA. .,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
40
|
Lukač N, Katavić V, Novak S, Šućur A, Filipović M, Kalajzić I, Grčević D, Kovačić N. What do we know about bone morphogenetic proteins and osteochondroprogenitors in inflammatory conditions? Bone 2020; 137:115403. [PMID: 32371019 DOI: 10.1016/j.bone.2020.115403] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/10/2020] [Accepted: 04/28/2020] [Indexed: 02/07/2023]
Abstract
Osteochondroprogenitors are crucial for embryonic bone development and postnatal processes such as bone repair in response to fracture injury, and their dysfunction may contribute to insufficient repair of structural damage in inflammatory arthritides. In the fracture healing, the early inflammatory phase is crucial for normal callus development and new bone formation. This process involves a complex interplay of many molecules and cell types, responsible for recruitment, expansion and differentiation of osteochondroprogenitor populations. In inflammatory arthritides, inflammation induces bone resorption and causes insufficient bone formation, which leads to local and systemic bone loss. While bone loss is a predominant feature in rheumatoid arthritis, inflammation also induces pathologic bone formation at enthesial sites in seronegative spondyloarthropathies. Bone morphogenetic proteins (BMP) are involved in cell proliferation, differentiation and apoptosis, and have fundamental roles in maintenance of postnatal bone homeostasis. They are crucial regulators of the osteochondroprogenitor pool and drive their proliferation, differentiation, and lifespan during bone regeneration. In this review, we summarize the effects of inflammation on osteochondroprogenitor populations during fracture repair and in inflammatory arthritides, with special focus on inflammation-mediated modulation of BMP signaling. We also present data in which we describe a population of murine synovial osteochondroprogenitor cells, which are reduced in arthritis, and characterize their expression of genes involved in regulation of bone homeostasis, emphasizing the up-regulation of BMP pathways in early progenitor subset. Based on the presented data, it may be concluded that during an inflammatory response, innate immune cells induce osteochondroprogenitors by providing signals for their recruitment, by producing BMPs and other osteogenic factors for paracrine effects, and by secreting inflammatory cytokines that may positively regulate osteogenic pathways. On the other hand, inflammatory cells may secrete cytokines that interfere with osteogenic pathways, proapoptotic factors that reduce the pool of osteochondroprogenitor cells, as well as BMP and Wnt antagonists. The net effect is strongly context-dependent and influenced by the local milieu of cells, cytokines, and growth factors. Further elucidation of the interplay between inflammatory signals and BMP-mediated bone formation may provide valuable tools for therapeutic targeting.
Collapse
Affiliation(s)
- Nina Lukač
- Laboratory for Molecular Immunology, University of Zagreb School of Medicine, Zagreb, Croatia; Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Vedran Katavić
- Laboratory for Molecular Immunology, University of Zagreb School of Medicine, Zagreb, Croatia; Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Sanja Novak
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT, USA
| | - Alan Šućur
- Laboratory for Molecular Immunology, University of Zagreb School of Medicine, Zagreb, Croatia; Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Maša Filipović
- Laboratory for Molecular Immunology, University of Zagreb School of Medicine, Zagreb, Croatia; Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Ivo Kalajzić
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT, USA
| | - Danka Grčević
- Laboratory for Molecular Immunology, University of Zagreb School of Medicine, Zagreb, Croatia; Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Nataša Kovačić
- Laboratory for Molecular Immunology, University of Zagreb School of Medicine, Zagreb, Croatia; Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia.
| |
Collapse
|
41
|
Di Pietro L, Barba M, Prampolini C, Ceccariglia S, Frassanito P, Vita A, Guadagni E, Bonvissuto D, Massimi L, Tamburrini G, Parolini O, Lattanzi W. GLI1 and AXIN2 Are Distinctive Markers of Human Calvarial Mesenchymal Stromal Cells in Nonsyndromic Craniosynostosis. Int J Mol Sci 2020; 21:E4356. [PMID: 32575385 PMCID: PMC7352200 DOI: 10.3390/ijms21124356] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 06/16/2020] [Indexed: 12/20/2022] Open
Abstract
All skeletal bones house osteogenic stem cell niches, in which mesenchymal stromal cells (MSC) provide progenitors for tissue growth and regeneration. They have been widely studied in long bones formed through endochondral ossification. Limited information is available on the composition of the osteogenic niche in flat bones (i.e., skull vault bones) that develop through direct membranous ossification. Craniosynostosis (CS) is a congenital craniofacial defect due to the excessive and premature ossification of skull vault sutures. This study aimed at analysing the expression of GLI1, AXIN2 and THY1 in the context of the human skull vault, using nonsyndromic forms of CS (NCS) as a model to test their functional implication in the aberrant osteogenic process. The expression of selected markers was studied in NCS patients' calvarial bone specimens, to assess the in vivo location of cells, and in MSC isolated thereof. The marker expression profile was analysed during in vitro osteogenic differentiation to validate the functional implication. Our results show that GLI1 and AXIN2 are expressed in periosteal and endosteal locations within the osteogenic niche of human calvarial bones. Their expression is higher in MSC isolated from calvarial bones than in those isolated from long bones and tends to decrease upon osteogenic commitment and differentiation. In particular, AXIN2 expression was lower in cells isolated from prematurely fused sutures than in those derived from patent sutures of NCS patients. This suggests that AXIN2 could reasonably represent a marker for the stem cell population that undergoes depletion during the premature ossification process occurring in CS.
Collapse
Affiliation(s)
- Lorena Di Pietro
- Dipartimento Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.D.P.); (M.B.); (S.C.); (A.V.); (E.G.); (O.P.)
| | - Marta Barba
- Dipartimento Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.D.P.); (M.B.); (S.C.); (A.V.); (E.G.); (O.P.)
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (P.F.); (D.B.); (L.M.); (G.T.)
| | - Chiara Prampolini
- Dipartimento Testa-Collo e Organi di Senso, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Sabrina Ceccariglia
- Dipartimento Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.D.P.); (M.B.); (S.C.); (A.V.); (E.G.); (O.P.)
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (P.F.); (D.B.); (L.M.); (G.T.)
| | - Paolo Frassanito
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (P.F.); (D.B.); (L.M.); (G.T.)
| | - Alessia Vita
- Dipartimento Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.D.P.); (M.B.); (S.C.); (A.V.); (E.G.); (O.P.)
| | - Enrico Guadagni
- Dipartimento Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.D.P.); (M.B.); (S.C.); (A.V.); (E.G.); (O.P.)
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (P.F.); (D.B.); (L.M.); (G.T.)
| | - Davide Bonvissuto
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (P.F.); (D.B.); (L.M.); (G.T.)
- Dipartimento Neuroscienze, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Luca Massimi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (P.F.); (D.B.); (L.M.); (G.T.)
- Dipartimento Neuroscienze, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Gianpiero Tamburrini
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (P.F.); (D.B.); (L.M.); (G.T.)
- Dipartimento Neuroscienze, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Ornella Parolini
- Dipartimento Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.D.P.); (M.B.); (S.C.); (A.V.); (E.G.); (O.P.)
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (P.F.); (D.B.); (L.M.); (G.T.)
| | - Wanda Lattanzi
- Dipartimento Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.D.P.); (M.B.); (S.C.); (A.V.); (E.G.); (O.P.)
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (P.F.); (D.B.); (L.M.); (G.T.)
| |
Collapse
|
42
|
Desterke C, Petit L, Sella N, Chevallier N, Cabeli V, Coquelin L, Durand C, Oostendorp RAJ, Isambert H, Jaffredo T, Charbord P. Inferring Gene Networks in Bone Marrow Hematopoietic Stem Cell-Supporting Stromal Niche Populations. iScience 2020; 23:101222. [PMID: 32535025 PMCID: PMC7300160 DOI: 10.1016/j.isci.2020.101222] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 03/19/2020] [Accepted: 05/27/2020] [Indexed: 02/07/2023] Open
Abstract
The cardinal property of bone marrow (BM) stromal cells is their capacity to contribute to hematopoietic stem cell (HSC) niches by providing mediators assisting HSC functions. In this study we first contrasted transcriptomes of stromal cells at different developmental stages and then included large number of HSC-supportive and non-supportive samples. Application of a combination of algorithms, comprising one identifying reliable paths and potential causative relationships in complex systems, revealed gene networks characteristic of the BM stromal HSC-supportive capacity and of defined niche populations of perivascular cells, osteoblasts, and mesenchymal stromal cells. Inclusion of single-cell transcriptomes enabled establishing for the perivascular cell subset a partially oriented graph of direct gene-to-gene interactions. As proof of concept we showed that R-spondin-2, expressed by the perivascular subset, synergized with Kit ligand to amplify ex vivo hematopoietic precursors. This study by identifying classifiers and hubs constitutes a resource to unravel candidate BM stromal mediators. A correlation network with predictor genes for the BM HSPC-supportive stromal niche An information theoretic network for the supportive perivascular stromal niche Wnt facilitator Rspo2 together with SCF to amplify ex vivo hematopoietic precursors Resource combining bioinformatics algorithms to search for novel stromal mediators
Collapse
Affiliation(s)
| | - Laurence Petit
- Sorbonne Université, UPMC Université Paris 06, IBPS, CNRS UMR7622, Inserm U 1156, Laboratoire de Biologie du Développement; Paris 75005, France
| | - Nadir Sella
- Institut Curie, PSL Research University, CNRS UMR168, Paris, France
| | - Nathalie Chevallier
- IMRB U955-E10, INSERM, Unité d'Ingenierie et de Thérapie Cellulaire- EFS, Université Paris-EST, Créteil, France
| | - Vincent Cabeli
- Institut Curie, PSL Research University, CNRS UMR168, Paris, France
| | - Laura Coquelin
- IMRB U955-E10, INSERM, Unité d'Ingenierie et de Thérapie Cellulaire- EFS, Université Paris-EST, Créteil, France
| | - Charles Durand
- Sorbonne Université, UPMC Université Paris 06, IBPS, CNRS UMR7622, Inserm U 1156, Laboratoire de Biologie du Développement; Paris 75005, France
| | - Robert A J Oostendorp
- Clinic and Polyclinic for Internal Medicine III, Klinikum Rechts der Isar, Technical University Munich, Munich, Germany
| | - Hervé Isambert
- Institut Curie, PSL Research University, CNRS UMR168, Paris, France
| | - Thierry Jaffredo
- Sorbonne Université, UPMC Université Paris 06, IBPS, CNRS UMR7622, Inserm U 1156, Laboratoire de Biologie du Développement; Paris 75005, France
| | - Pierre Charbord
- Sorbonne Université, UPMC Université Paris 06, IBPS, CNRS UMR7622, Inserm U 1156, Laboratoire de Biologie du Développement; Paris 75005, France.
| |
Collapse
|
43
|
Xu GP, Zhang XF, Sun L, Chen EM. Current and future uses of skeletal stem cells for bone regeneration. World J Stem Cells 2020; 12:339-350. [PMID: 32547682 PMCID: PMC7280866 DOI: 10.4252/wjsc.v12.i5.339] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 04/07/2020] [Accepted: 04/18/2020] [Indexed: 02/06/2023] Open
Abstract
The postnatal skeleton undergoes growth, modeling, and remodeling. The human skeleton is a composite of diverse tissue types, including bone, cartilage, fat, fibroblasts, nerves, blood vessels, and hematopoietic cells. Fracture nonunion and bone defects are among the most challenging clinical problems in orthopedic trauma. The incidence of nonunion or bone defects following fractures is increasing. Stem and progenitor cells mediate homeostasis and regeneration in postnatal tissue, including bone tissue. As multipotent stem cells, skeletal stem cells (SSCs) have a strong effect on the growth, differentiation, and repair of bone regeneration. In recent years, a number of important studies have characterized the hierarchy, differential potential, and bone formation of SSCs. Here, we describe studies on and applications of SSCs and/or mesenchymal stem cells for bone regeneration.
Collapse
Affiliation(s)
- Guo-Ping Xu
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Xiang-Feng Zhang
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Lu Sun
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, MA 02115, United States
| | - Er-Man Chen
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| |
Collapse
|
44
|
Tournaire G, Stegen S, Giacomini G, Stockmans I, Moermans K, Carmeliet G, van Gastel N. Nestin-GFP transgene labels skeletal progenitors in the periosteum. Bone 2020; 133:115259. [PMID: 32036051 DOI: 10.1016/j.bone.2020.115259] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/20/2020] [Accepted: 02/01/2020] [Indexed: 02/07/2023]
Abstract
The periosteum is critical for bone repair and contains skeletal stem cells (SSCs), but these cells are still poorly characterized. In the bone marrow, cells expressing the Nes-GFP transgene have been described to be SSCs. Here, we investigated whether Nes-GFP expression also typifies SSCs in the periosteum. We show that in adult mice, Nes-GFP cells are present in the periosteum and localize closely to blood vessels, but periosteal Nes-GFP cells express SSC and progenitor markers differently compared to Nes-GFP cells in the bone marrow. Periosteal Nes-GFP cells show in vitro clonogenicity and tri-lineage differentiation potential and they can form bone in vivo. Shortly after fracture, they start to proliferate and they contribute to the osteoblast pool during the repair process. However, periosteal Nes-GFP cells are not slow dividing nor self-renewing in vivo. These results indicate that in adult mice, periosteal Nes-GFP expressing cells are skeletal progenitors rather than true SSCs, and they participate in the fracture healing process.
Collapse
Affiliation(s)
- Guillaume Tournaire
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Steve Stegen
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Greta Giacomini
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Ingrid Stockmans
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Karen Moermans
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Geert Carmeliet
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium.
| | - Nick van Gastel
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| |
Collapse
|
45
|
Wang D, Gilbert JR, Zhang X, Zhao B, Ker DFE, Cooper GM. Calvarial Versus Long Bone: Implications for Tailoring Skeletal Tissue Engineering. TISSUE ENGINEERING PART B-REVIEWS 2019; 26:46-63. [PMID: 31588853 DOI: 10.1089/ten.teb.2018.0353] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Tissue-engineered graft substitutes have shown great potential to treat large bone defects. While we usually assume that therapeutic approaches developed for appendicular bone healing could be similarly translated for application in craniofacial reconstruction and vice versa, this is not necessarily accurate. In addition to those more well-known healing-associated factors, such as age, lifestyle (e.g., nutrition and smoking), preexisting disease (e.g., diabetes), medication, and poor blood supply, the developmental origins and surrounding tissue of the wound sites can largely affect the fracture healing outcome as well as designed treatments. Therefore, the strategies developed for long bone fracture repair might not be suitable or directly applicable to skull bone repair. In this review, we discuss aspects of development, healing process, structure, and tissue engineering considerations between calvarial and long bones to assist in designing the tailored bone repair strategies. Impact Statement We summarized, in this review, the existing body of knowledge between long bone and calvarial bone with regard to their development and healing, tissue structure, and consideration of current tissue engineering strategies. By highlighting their similarities and differences, we propose that tailored tissue engineering strategies, such as scaffold features, growth factor usage, and the source of cells for tissue or region-specific bone repair, are necessary to ensure an optimized healing outcome.
Collapse
Affiliation(s)
- Dan Wang
- Department of Stomatology, Tenth People's Hospital of Tongji University, Shanghai, China.,Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - James R Gilbert
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania.,McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Xu Zhang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Bingkun Zhao
- Department of Stomatology, Tenth People's Hospital of Tongji University, Shanghai, China.,Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Dai Fei Elmer Ker
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Gregory M Cooper
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Oral Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
46
|
Expanded skeletal stem and progenitor cells promote and participate in induced bone regeneration at subcritical BMP-2 dose. Biomaterials 2019; 217:119278. [DOI: 10.1016/j.biomaterials.2019.119278] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 06/11/2019] [Accepted: 06/11/2019] [Indexed: 01/05/2023]
|
47
|
Ambrosi TH, Longaker MT, Chan CKF. A Revised Perspective of Skeletal Stem Cell Biology. Front Cell Dev Biol 2019; 7:189. [PMID: 31572721 PMCID: PMC6753172 DOI: 10.3389/fcell.2019.00189] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 08/23/2019] [Indexed: 12/20/2022] Open
Abstract
Bone-related maladies are a major health burden on modern society. Loss of skeletal integrity and regeneration capacity through aging, obesity, and disease follows from a detrimental shift in bone formation and resorption dynamics. Targeting tissue-resident adult stem cells offers a potentially innovative paradigm in the development of therapeutic strategies against organ dysfunction. While the essential role of skeletal stem cells (SSCs) for development, growth, and maintenance of the skeleton has been generally established, a common consensus on the exact identity and definition of a pure bona fide SSC population remains elusive. The controversies stem from conflicting results between different approaches and criteria for isolation, detection, and functional evaluation; along with the interchangeable usage of the terms SSC and "mesenchymal stromal/stem cell (MSC)". A great number of prospective bone-forming stem cell populations have been reported with various characteristic markers, often describing overlapping cell populations with widely unexplored heterogeneity, species specificity, and distribution at distinct skeletal sites, bone regions, and microenvironments, thereby creating confusion that may complicate future advances in the field. In this review, we examine the state-of-the-art knowledge of SSC biology and try to establish a common ground for the definition and terminology of specific bone-resident stem cells. We also discuss recent advances in the identification of highly purified SSCs, which will allow detailed interrogation of SSC diversity and regulation at the single-cell level.
Collapse
Affiliation(s)
- Thomas H Ambrosi
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Michael T Longaker
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States.,Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Charles K F Chan
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States.,Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
48
|
Zhu F, Feng M, Sinha R, Murphy MP, Luo F, Kao KS, Szade K, Seita J, Weissman IL. The GABA receptor GABRR1 is expressed on and functional in hematopoietic stem cells and megakaryocyte progenitors. Proc Natl Acad Sci U S A 2019; 116:18416-18422. [PMID: 31451629 PMCID: PMC6744911 DOI: 10.1073/pnas.1906251116] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
GABRR1 is a rho subunit receptor of GABA, the major inhibitory neurotransmitter in the mammalian brain. While most investigations of its function focused on the nervous system, its regulatory role in hematopoiesis has not been reported. In this study, we found GABRR1 is mainly expressed on subsets of human and mouse hematopoietic stem cells (HSCs) and megakaryocyte progenitors (MkPs). GABRR1-negative (GR-) HSCs led to higher donor-derived hematopoietic chimerism than GABRR1-positive (GR+) HSCs. GR+ but not GR- HSCs and MkPs respond to GABA in patch clamp studies. Inhibition of GABRR1 via genetic knockout or antagonists inhibited MkP differentiation and reduced platelet numbers in blood. Overexpression of GABRR1 or treatment with agonists significantly promoted MkP generation and megakaryocyte colonies. Thus, this study identifies a link between the neural and hematopoietic systems and opens up the possibility of manipulating GABA signaling for platelet-required clinical applications.
Collapse
Affiliation(s)
- Fangfang Zhu
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305;
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Mingye Feng
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010
| | - Rahul Sinha
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Matthew Philip Murphy
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, CA 94305
- Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery Division, Stanford University School of Medicine, Stanford, CA 94305
| | - Fujun Luo
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305
| | - Kevin S Kao
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Krzysztof Szade
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Jun Seita
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Irving L Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305;
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
49
|
Wasnik S, Lakhan R, Baylink DJ, Rundle CH, Xu Y, Zhang J, Qin X, Lau KHW, Carreon EE, Tang X. Cyclooxygenase 2 augments osteoblastic but suppresses chondrocytic differentiation of CD90 + skeletal stem cells in fracture sites. SCIENCE ADVANCES 2019; 5:eaaw2108. [PMID: 31392271 PMCID: PMC6669009 DOI: 10.1126/sciadv.aaw2108] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 06/21/2019] [Indexed: 05/07/2023]
Abstract
Cyclooxygenase 2 (COX-2) is essential for normal tissue repair. Although COX-2 is known to enhance the differentiation of mesenchymal stem cells (MSCs), how COX-2 regulates MSC differentiation into different tissue-specific progenitors to promote tissue repair remains unknown. Because it has been shown that COX-2 is critical for normal bone repair and local COX-2 overexpression in fracture sites accelerates fracture repair, this study aimed to determine the MSC subsets that are targeted by COX-2. We showed that CD90+ mouse skeletal stem cells (mSSCs; i.e., CD45-Tie2-AlphaV+ MSCs) were selectively recruited by macrophage/monocyte chemoattractant protein 1 into fracture sites following local COX-2 overexpression. In addition, local COX-2 overexpression augmented osteoblast differentiation and suppressed chondrocyte differentiation in CD90+ mSSCs, which depended on canonical WNT signaling. CD90 depletion data demonstrated that local COX-2 overexpression targeted CD90+ mSSCs to accelerate fracture repair. In conclusion, CD90+ mSSCs are promising targets for the acceleration of bone repair.
Collapse
Affiliation(s)
- Samiksha Wasnik
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Ram Lakhan
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - David J. Baylink
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Charles H. Rundle
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial Veterans Affairs Medical Center, Loma Linda, CA, USA
| | - Yi Xu
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Jintao Zhang
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, USA
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Henan, China
| | - Xuezhong Qin
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, USA
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial Veterans Affairs Medical Center, Loma Linda, CA, USA
| | - Kin-Hing William Lau
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, USA
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial Veterans Affairs Medical Center, Loma Linda, CA, USA
| | - Edmundo E. Carreon
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Xiaolei Tang
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, USA
- Corresponding author.
| |
Collapse
|
50
|
Chan CKF, Gulati GS, Sinha R, Tompkins JV, Lopez M, Carter AC, Ransom RC, Reinisch A, Wearda T, Murphy M, Brewer RE, Koepke LS, Marecic O, Manjunath A, Seo EY, Leavitt T, Lu WJ, Nguyen A, Conley SD, Salhotra A, Ambrosi TH, Borrelli MR, Siebel T, Chan K, Schallmoser K, Seita J, Sahoo D, Goodnough H, Bishop J, Gardner M, Majeti R, Wan DC, Goodman S, Weissman IL, Chang HY, Longaker MT. Identification of the Human Skeletal Stem Cell. Cell 2019; 175:43-56.e21. [PMID: 30241615 DOI: 10.1016/j.cell.2018.07.029] [Citation(s) in RCA: 418] [Impact Index Per Article: 69.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 01/16/2018] [Accepted: 07/20/2018] [Indexed: 12/14/2022]
Abstract
Stem cell regulation and hierarchical organization of human skeletal progenitors remain largely unexplored. Here, we report the isolation of a self-renewing and multipotent human skeletal stem cell (hSSC) that generates progenitors of bone, cartilage, and stroma, but not fat. Self-renewing and multipotent hSSCs are present in fetal and adult bones and can also be derived from BMP2-treated human adipose stroma (B-HAS) and induced pluripotent stem cells (iPSCs). Gene expression analysis of individual hSSCs reveals overall similarity between hSSCs obtained from different sources and partially explains skewed differentiation toward cartilage in fetal and iPSC-derived hSSCs. hSSCs undergo local expansion in response to acute skeletal injury. In addition, hSSC-derived stroma can maintain human hematopoietic stem cells (hHSCs) in serum-free culture conditions. Finally, we combine gene expression and epigenetic data of mouse skeletal stem cells (mSSCs) and hSSCs to identify evolutionarily conserved and divergent pathways driving SSC-mediated skeletogenesis. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Charles K F Chan
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA.
| | - Gunsagar S Gulati
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Rahul Sinha
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | | | - Michael Lopez
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Ava C Carter
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
| | - Ryan C Ransom
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Andreas Reinisch
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Taylor Wearda
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Matthew Murphy
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Rachel E Brewer
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Lauren S Koepke
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Owen Marecic
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Anoop Manjunath
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Eun Young Seo
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Tripp Leavitt
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Wan-Jin Lu
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Allison Nguyen
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Stephanie D Conley
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Ankit Salhotra
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Thomas H Ambrosi
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Mimi R Borrelli
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Taylor Siebel
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Karen Chan
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Katharina Schallmoser
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Jun Seita
- Medical Sciences Innovation Hub Program, RIKEN, Tokyo 103-0027, Japan
| | - Debashis Sahoo
- Departments of Pediatrics and Computer Science and Engineering, University of California San Diego, San Diego, CA 92093, USA
| | - Henry Goodnough
- Department of Orthopedic Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Julius Bishop
- Department of Orthopedic Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Michael Gardner
- Department of Orthopedic Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Ravindra Majeti
- Department of Medicine, Division of Hematology, Stanford Medicine, Stanford, CA 94305, USA
| | - Derrick C Wan
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Stuart Goodman
- Department of Orthopedic Surgery, Stanford Medicine, Stanford, CA 94305, USA
| | - Irving L Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
| | - Michael T Longaker
- Department of Surgery, Stanford Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford Medicine, Stanford, CA 94305, USA.
| |
Collapse
|