The enhancer landscape predetermines the skeletal regeneration capacity of stromal cells

Multipotent stromal cells are considered attractive sources for cell therapy and tissue engineering. Despite numerous experimental and clinical studies, broad application of stromal cell therapeutics is not yet emerging. A major challenge is the functional diversity of available cell sources. Here, we investigated the regenerative potential of clinically relevant human stromal cells from bone marrow (BMSCs), white adipose tissue, and umbilical cord compared with mature chondrocytes and skin fibroblasts in vitro and in vivo. Although all stromal cell types could express transcription factors related to endochondral ossification, only BMSCs formed cartilage discs in vitro that fully regenerated critical-size femoral defects after transplantation into mice. We identified cell type–specific epigenetic landscapes as the underlying molecular mechanism controlling transcriptional stromal differentiation networks. Binding sites of commonly expressed transcription factors in the enhancer and promoter regions of ossification-related genes, including Runt and bZIP families, were accessible only in BMSCs but not in extraskeletal stromal cells. This suggests an epigenetically predetermined differentiation potential depending on cell origin that allows common transcription factors to trigger distinct organ-specific transcriptional programs, facilitating forward selection of regeneration-competent cell sources. Last, we demonstrate that viable human BMSCs initiated defect healing through the secretion of osteopontin and contributed to transient mineralized bone hard callus formation after transplantation into immunodeficient mice, which was eventually replaced by murine recipient bone during final tissue remodeling. Description Stromal cells vary in their tissue-specific differentiation potential because of their differing epigenetic enhancer signatures. Enhancing bone ossification Musculoskeletal tissue engineering approaches require a reliable source of precursor cells. Hochmann et al. report that despite having some transcriptional similarities, the inherent differentiation potential of different stromal cell types is constrained by their enhancer landscapes. In contrast to stromal cells derived from white adipose tissue or umbilical cord, only bone marrow–derived cells had an epigenetic signature permitting their differentiation into functional hypertrophic cartilage discs capable of ameliorating a critical-size bone defect model after transplantation into mice. These findings have ramifications for the development of regenerative cell–based musculoskeletal therapies. —CAC

[1]  Antonina Hafner,et al.  The spatial organization of transcriptional control , 2022, Nature reviews. Genetics.

[2]  D. Strunk,et al.  Batch Effects during Human Bone Marrow Stromal Cell Propagation Prevail Donor Variation and Culture Duration: Impact on Genotype, Phenotype and Function , 2022, Cells.

[3]  D. Strunk,et al.  Self-assembly of differentiated progenitor cells facilitates spheroid human skin organoid formation and planar skin regeneration , 2021, Theranostics.

[4]  J. Schulte,et al.  Discovery of Spatial Peptide Signatures for Neuroblastoma Risk Assessment by MALDI Mass Spectrometry Imaging , 2021, Cancers.

[5]  S. Mandrup,et al.  Transcriptional networks controlling stromal cell differentiation , 2021, Nature Reviews Molecular Cell Biology.

[6]  D. Pei,et al.  Challenges and advances in clinical applications of mesenchymal stromal cells , 2021, Journal of Hematology & Oncology.

[7]  Qingfeng Li,et al.  Bone defect reconstruction via endochondral ossification: A developmental engineering strategy , 2020, Journal of tissue engineering.

[8]  A. Arthur,et al.  Clinical Application of Bone Marrow Mesenchymal Stem/Stromal Cells to Repair Skeletal Tissue , 2020, International journal of molecular sciences.

[9]  M. Longaker,et al.  Mechanisms of bone development and repair , 2020, Nature Reviews Molecular Cell Biology.

[10]  G. Duda,et al.  Mesenchymal stromal cell and bone marrow concentrate therapies for musculoskeletal indications: a concise review of current literature , 2020, Molecular Biology Reports.

[11]  M. Lenardo,et al.  A guide to cancer immunotherapy: from T cell basic science to clinical practice , 2020, Nature Reviews Immunology.

[12]  M. Lenardo,et al.  A guide to cancer immunotherapy: from T cell basic science to clinical practice , 2020, Nature Reviews Immunology.

[13]  F. Luyten,et al.  Developmentally Engineered Callus Organoid Bioassemblies Exhibit Predictive In Vivo Long Bone Healing , 2019, Advanced science.

[14]  M. Tognon,et al.  Adult Stem Cells for Bone Regeneration and Repair , 2019, Front. Cell Dev. Biol..

[15]  J. Chimal-Monroy,et al.  Understanding the Cellular and Molecular Mechanisms That Control Early Cell Fate Decisions During Appendicular Skeletogenesis , 2019, Front. Genet..

[16]  S. Stewart Fracture Non-Union: A Review of Clinical Challenges and Future Research Needs , 2019, Malaysian orthopaedic journal.

[17]  Eamon J. Sheehy,et al.  Biomaterial-based endochondral bone regeneration: a shift from traditional tissue engineering paradigms to developmentally inspired strategies , 2019, Materials today. Bio.

[18]  G. Duda,et al.  Experience in the Adaptive Immunity Impacts Bone Homeostasis, Remodeling, and Healing , 2019, Front. Immunol..

[19]  Anders K. Haakonsson,et al.  Osteogenesis depends on commissioning of a network of stem cell transcription factors that act as repressors of adipogenesis , 2019, Nature Genetics.

[20]  R. Marcucio,et al.  Cellular biology of fracture healing , 2018, Journal of orthopaedic research : official publication of the Orthopaedic Research Society.

[21]  B. Gabbe,et al.  Incidence, Costs and Predictors of Non-Union, Delayed Union and Mal-Union Following Long Bone Fracture , 2018, International journal of environmental research and public health.

[22]  H. Leemhuis,et al.  A biomaterial with a channel-like pore architecture induces endochondral healing of bone defects , 2018, Nature Communications.

[23]  D. Sipp,et al.  Clear up this stem-cell mess , 2018, Nature.

[24]  Rachel E. Brewer,et al.  Identification of the Human Skeletal Stem Cell , 2018, Cell.

[25]  J. H. Kobarg,et al.  Unraveling local tissue changes within severely injured skeletal muscles in response to MSC-based intervention using MALDI Imaging mass spectrometry , 2018, Scientific Reports.

[26]  I. Martin,et al.  Ectopic bone formation by aggregated mesenchymal stem cells from bone marrow and adipose tissue: A comparative study , 2018, Journal of tissue engineering and regenerative medicine.

[27]  S. Buratowski,et al.  Determinants of Histone H3K4 Methylation Patterns. , 2017, Molecular cell.

[28]  A. Kamali,et al.  Role of Mesenchymal Stem Cells in Bone Regenerative Medicine: What Is the Evidence? , 2017, Cells Tissues Organs.

[29]  P. Robey,et al.  “Mesenchymal stem cells”: fact or fiction, and implications in their therapeutic use , 2017, F1000Research.

[30]  Giulio Cossu,et al.  No Identical “Mesenchymal Stem Cells” at Different Times and Sites: Human Committed Progenitors of Distinct Origin and Differentiation Potential Are Incorporated as Adventitial Cells in Microvessels , 2016, Stem cell reports.

[31]  A. Reinisch,et al.  A humanized bone marrow ossicle xenotransplantation model enables improved engraftment of healthy and leukemic human hematopoietic cells , 2016, Nature Medicine.

[32]  A. Ignatius,et al.  Fracture Healing Is Delayed in Immunodeficient NOD/scid‑IL2Rγcnull Mice , 2016, PloS one.

[33]  Christina S. Leslie,et al.  Early enhancer establishment and regulatory locus complexity shape transcriptional programs in hematopoietic differentiation , 2015, Nature Genetics.

[34]  Cory J. Berkland,et al.  Endochondral ossification for enhancing bone regeneration: converging native extracellular matrix biomaterials and developmental engineering in vivo. , 2015, Tissue engineering. Part B, Reviews.

[35]  F. O'Valle,et al.  Clinical Application of Mesenchymal Stem Cells and Novel Supportive Therapies for Oral Bone Regeneration , 2015, BioMed research international.

[36]  I. Weissman,et al.  Epigenetic and in vivo comparison of diverse MSC sources reveals an endochondral signature for human hematopoietic niche formation. , 2015, Blood.

[37]  Howard Y. Chang,et al.  ATAC‐seq: A Method for Assaying Chromatin Accessibility Genome‐Wide , 2015, Current protocols in molecular biology.

[38]  B. Olsen,et al.  Bone development. , 2015, Bone.

[39]  R. Serra,et al.  Runx2 Regulates Endochondral Ossification Through Control of Chondrocyte Proliferation and Differentiation , 2014, Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research.

[40]  G. Logroscino,et al.  Bone substitutes in orthopaedic surgery: from basic science to clinical practice , 2014, Journal of Materials Science: Materials in Medicine.

[41]  C. Wells,et al.  YuGene: a simple approach to scale gene expression data derived from different platforms for integrated analyses. , 2014, Genomics.

[42]  Zhi Wei,et al.  Generation of folliculogenic human epithelial stem cells from induced pluripotent stem cells , 2014, Nature Communications.

[43]  J. Triffitt,et al.  Faculty Opinions recommendation of The meaning, the sense and the significance: translating the science of mesenchymal stem cells into medicine. , 2013 .

[44]  Navrag B. Singh,et al.  Terminally Differentiated CD8+ T Cells Negatively Affect Bone Regeneration in Humans , 2013, Science Translational Medicine.

[45]  Xu Cao,et al.  The meaning, the sense and the significance: translating the science of mesenchymal stem cells into medicine , 2013, Nature Medicine.

[46]  R. Zhao,et al.  JOURNAL OF HEMATOLOGY & ONCOLOGY REVIEW Open Access Clinical applications of mesenchymal stem cells , 2022 .

[47]  Orlando Guntinas-Lichius,et al.  MALDI-imaging segmentation is a powerful tool for spatial functional proteomic analysis of human larynx carcinoma , 2012, Journal of Cancer Research and Clinical Oncology.

[48]  R. Young,et al.  Histone H3K27ac separates active from poised enhancers and predicts developmental state , 2010, Proceedings of the National Academy of Sciences.

[49]  Theodore Alexandrov,et al.  Spatial segmentation of imaging mass spectrometry data with edge-preserving image denoising and clustering. , 2010, Journal of proteome research.

[50]  Ivan Martin,et al.  Recapitulation of endochondral bone formation using human adult mesenchymal stem cells as a paradigm for developmental engineering , 2010, Proceedings of the National Academy of Sciences.

[51]  Petros Lenas,et al.  Developmental engineering: a new paradigm for the design and manufacturing of cell-based products. Part I: from three-dimensional cell growth to biomimetics of in vivo development. , 2009, Tissue engineering. Part B, Reviews.

[52]  A. Reinisch,et al.  Humanized large-scale expanded endothelial colony-forming cells function in vitro and in vivo. , 2009, Blood.

[53]  A. Reinisch,et al.  Rapid large-scale expansion of functional mesenchymal stem cells from unmanipulated bone marrow without animal serum. , 2008, Tissue engineering. Part C, Methods.

[54]  J. Rossant,et al.  Bone sialoprotein plays a functional role in bone formation and osteoclastogenesis , 2008, The Journal of experimental medicine.

[55]  P. Bianco,et al.  Mesenchymal stem cells: revisiting history, concepts, and assays. , 2008, Cell stem cell.

[56]  T. Hardingham,et al.  Chondrogenic Differentiation of Human Bone Marrow Stem Cells in Transwell Cultures: Generation of Scaffold‐Free Cartilage , 2007, Stem cells.

[57]  Maurilio Marcacci,et al.  Stem cells associated with macroporous bioceramics for long bone repair: 6- to 7-year outcome of a pilot clinical study. , 2007, Tissue engineering.

[58]  A. Barbero,et al.  Visual histological grading system for the evaluation of in vitro-generated neocartilage. , 2006, Tissue engineering.

[59]  P. Kasten,et al.  Platelet-rich plasma improves expansion of human mesenchymal stem cells and retains differentiation capacity and in vivo bone formation in calcium phosphate ceramics , 2006, Platelets.

[60]  R. Handgretinger,et al.  Animal serum-free culture conditions for isolation and expansion of multipotent mesenchymal stromal cells from human BM. , 2006, Cytotherapy.

[61]  D. Prockop,et al.  Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. , 2006, Cytotherapy.

[62]  R Cancedda,et al.  Repair of large bone defects with the use of autologous bone marrow stromal cells. , 2001, The New England journal of medicine.

[63]  C Perka,et al.  Segmental bone repair by tissue-engineered periosteal cell transplants with bioresorbable fleece and fibrin scaffolds in rabbits. , 2000, Biomaterials.

[64]  Evon M. O. Abu-Taieh,et al.  Comparative Study , 2020, Definitions.

[65]  E. Ogata Studies on parathyroid hormone, vitamin D and pseudohypoparathyroidism: an example of trail from basic science to clinical practice. , 1989, Japanese journal of medicine.