305 results on '"Pamela Gehron Robey"'
Search Results
2. Characterisation of ovine bone marrow-derived stromal cells (oBMSC) and evaluation of chondrogenically induced micro-pellets for cartilage tissue repair in vivo
- Author
-
Kathryn Futrega, Ena Music, Ross Crawford, Pamela Gehron Robey, Stan Gronthos, Siamak Saifzadeh, Travis J. Klein, and Michael R. Doran
- Subjects
Cartilage, Articular ,IBMX ,Stromal cell ,Medicine (miscellaneous) ,Bone Marrow Cells ,Pilot Projects ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,lcsh:Biochemistry ,chemistry.chemical_compound ,Chondrocytes ,Bone Marrow ,Osteoarthritis ,medicine ,Animals ,lcsh:QD415-436 ,Micro-pellet ,Cells, Cultured ,lcsh:R5-920 ,Sheep ,Research ,Cartilage ,Mesenchymal stem cell ,Bone marrow stromal cells ,Cell Differentiation ,Hypertrophy ,Cell Biology ,Chondrogenesis ,Cell biology ,Oxygen ,medicine.anatomical_structure ,chemistry ,Adipogenesis ,Differentiation ,Molecular Medicine ,Mesenchymal stem cells ,Bone marrow ,Stem cell ,lcsh:Medicine (General) - Abstract
Abstract Bone marrow stromal cells (BMSC) show promise in cartilage repair, and sheep are the most common large animal pre-clinical model. Objective The objective of this study was to characterise ovine BMSC (oBMSC) in vitro, and to evaluate the capacity of chondrogenic micro-pellets manufactured from oBMSC or ovine articular chondrocytes (oACh) to repair osteochondral defects in sheep. Design oBMSC were characterised for surface marker expression using flow cytometry and evaluated for tri-lineage differentiation capacity. oBMSC micro-pellets were manufactured in a microwell platform, and chondrogenesis was compared at 2%, 5%, and 20% O2. The capacity of cartilage micro-pellets manufactured from oBMSC or oACh to repair osteochondral defects in adult sheep was evaluated in an 8-week pilot study. Results Expanded oBMSC were positive for CD44 and CD146 and negative for CD45. The common adipogenic induction ingredient, 3-Isobutyl-1-methylxanthine (IBMX), was toxic to oBMSC, but adipogenesis could be restored by excluding IBMX from the medium. BMSC chondrogenesis was optimal in a 2% O2 atmosphere. Micro-pellets formed from oBMSC or oACh appeared morphologically similar, but hypertrophic genes were elevated in oBMSC micro-pellets. While oACh micro-pellets formed cartilage-like repair tissue in sheep, oBMSC micro-pellets did not. Conclusion The sensitivity of oBMSC, compared to human BMSC, to IBMX in standard adipogenic assays highlights species-associated differences. Micro-pellets manufactured from oACh were more effective than micro-pellets manufactured from oBMSC in the repair of osteochondral defects in sheep. While oBMSC can be driven to form cartilage-like tissue in vitro, the effective use of these cells in cartilage repair will depend on the successful mitigation of hypertrophy and tissue integration.
- Published
- 2021
3. Review for 'Parathyroid hormone therapy for managing chronic hypoparathyroidism: a systematic review and meta‐analysis'
- Author
-
null Pamela Gehron Robey
- Published
- 2022
- Full Text
- View/download PDF
4. Recent updates on the biological basis of heterogeneity in bone marrow stromal cells/skeletal stem cells
- Author
-
Deepika, Arora and Pamela Gehron, Robey
- Abstract
Based on studies over the last several decades, the self-renewing skeletal lineages derived from bone marrow stroma could be an ideal source for skeletal tissue engineering. However, the markers for osteogenic precursors; i.e., bone marrowderived skeletal stem cells (SSCs), in association with other cells of the marrow stroma (bone marrow stromal cells, BMSCs) and their heterogeneous nature both in vivo and in vitro remain to be clarified. This review aims to highlight: i) the importance of distinguishing BMSCs/SSCs from other "mesenchymal stem/stromal cells", and ii) factors that are responsible for their heterogeneity, and how these factors impact on the differentiation potential of SSCs towards bone. The prospective role of SSC enrichment, their expansion and its impact on SSC phenotype is explored. Emphasis has also been given to emerging single cell RNA sequencing approaches in scrutinizing the unique population of SSCs within the BMSC population, along with their committed progeny. Understanding the factors involved in heterogeneity may help researchers to improvise their strategies to isolate, characterize and adopt best culture practices and source identification to develop standard operating protocols for developing reproducible stem cells grafts. However, more scientific understanding of the molecular basis of heterogeneity is warranted that may be obtained from the robust high-throughput functional transcriptomics of single cells or clonal populations.
- Published
- 2022
5. GsαR201C and estrogen reveal different subsets of bone marrow adiponectin expressing osteogenic cells
- Author
-
Biagio Palmisano, Rossella Labella, Samantha Donsante, Cristina Remoli, Emanuela Spica, Ilenia Coletta, Giorgia Farinacci, Michele Dello Spedale Venti, Isabella Saggio, Marta Serafini, Pamela Gehron Robey, Alessandro Corsi, and Mara Riminucci
- Subjects
Histology ,adiponectin ,Physiology ,Endocrinology, Diabetes and Metabolism ,fibrous dysplasia ,estrogen ,pericytes ,marrow progenitors ,bone marrow stroma ,BMSCs ,osteogenesis ,intramedullary bone ,GNAS - Abstract
The Gsα/cAMP signaling pathway mediates the effect of a variety of hormones and factors that regulate the homeostasis of the post-natal skeleton. Hence, the dysregulated activity of Gsα due to gain-of-function mutations (R201C/R201H) results in severe architectural and functional derangements of the entire bone/bone marrow organ. While the consequences of gain-of-function mutations of Gsα have been extensively investigated in osteoblasts and in bone marrow osteoprogenitor cells at various differentiation stages, their effect in adipogenically-committed bone marrow stromal cells has remained unaddressed. We generated a mouse model with expression of GsαR201C driven by the Adiponectin (Adq) promoter. Adq-GsαR201C mice developed a complex combination of metaphyseal, diaphyseal and cortical bone changes. In the metaphysis, GsαR201C caused an early phase of bone resorption followed by bone deposition. Metaphyseal bone formation was sustained by cells that were traced by Adq-Cre and eventually resulted in a high trabecular bone mass phenotype. In the diaphysis, GsαR201C, in combination with estrogen, triggered the osteogenic activity of Adq-Cre-targeted perivascular bone marrow stromal cells leading to intramedullary bone formation. Finally, consistent with the previously unnoticed presence of Adq-Cre-marked pericytes in intraosseous blood vessels, GsαR201C caused the development of a lytic phenotype that affected both cortical (increased porosity) and trabecular (tunneling resorption) bone. These results provide the first evidence that the Adq-cell network in the skeleton not only regulates bone resorption but also contributes to bone formation, and that the Gsα/cAMP pathway is a major modulator of both functions.
- Published
- 2022
6. Gsα
- Author
-
Biagio, Palmisano, Rossella, Labella, Samantha, Donsante, Cristina, Remoli, Emanuela, Spica, Ilenia, Coletta, Giorgia, Farinacci, Michele, Dello Spedale Venti, Isabella, Saggio, Marta, Serafini, Pamela Gehron, Robey, Alessandro, Corsi, and Mara, Riminucci
- Abstract
The Gsα/cAMP signaling pathway mediates the effect of a variety of hormones and factors that regulate the homeostasis of the post-natal skeleton. Hence, the dysregulated activity of Gsα due to gain-of-function mutations (R201C/R201H) results in severe architectural and functional derangements of the entire bone/bone marrow organ. While the consequences of gain-of-function mutations of Gsα have been extensively investigated in osteoblasts and in bone marrow osteoprogenitor cells at various differentiation stages, their effect in adipogenically-committed bone marrow stromal cells has remained unaddressed. We generated a mouse model with expression of Gsα
- Published
- 2021
7. Secreted frizzled related-protein 2 (Sfrp2) deficiency decreases adult skeletal stem cell function in mice
- Author
-
Yankel Gabet, Brian Sworder, Nathan J. Burbach, Byron W H Mui, Luis F de Castro, Kenn Holmbeck, Agnes D. Berendsen, Sergei A. Kuznetsov, Natasha Cherman, Pamela Gehron Robey, Matthew D. Phillips, and Kathryn Futrega
- Subjects
Frizzled ,endocrine system ,Histology ,Stromal cell ,QH301-705.5 ,Physiology ,Endocrinology, Diabetes and Metabolism ,Regeneration (biology) ,Mesenchymal stem cell ,Wnt signaling pathway ,LRP6 ,Biology ,Article ,Cell biology ,Bone quality and biomechanics ,RUNX2 ,stomatognathic system ,QP1-981 ,Biology (General) ,Stem cell ,Bone - Abstract
In a previous transcriptomic study of human bone marrow stromal cells (BMSCs, also known as bone marrow-derived “mesenchymal stem cells”), SFRP2 was highly over-represented in a subset of multipotent BMSCs (skeletal stem cells, SSCs), which recreate a bone/marrow organ in an in vivo ectopic bone formation assay. SFRPs modulate WNT signaling, which is essential to maintain skeletal homeostasis, but the specific role of SFRP2 in BMSCs/SSCs is unclear. Here, we evaluated Sfrp2 deficiency on BMSC/SSC function in models of skeletal organogenesis and regeneration. The skeleton of Sfrp2-deficient (KO) mice is overtly normal; but their BMSCs/SSCs exhibit reduced colony-forming efficiency, reflecting low SSC self-renewal/abundancy. Sfrp2 KO BMSCs/SSCs formed less trabecular bone than those from WT littermates in the ectopic bone formation assay. Moreover, regeneration of a cortical drilled hole defect was dramatically impaired in Sfrp2 KO mice. Sfrp2-deficient BMSCs/SSCs exhibited poor in vitro osteogenic differentiation as measured by Runx2 and Osterix expression and calcium accumulation. Interestingly, activation of the Wnt co-receptor, Lrp6, and expression of Wnt target genes, Axin2, C-myc and Cyclin D1, were reduced in Sfrp2-deficient BMSCs/SSCs. Addition of recombinant Sfrp2 restored most of these activities, suggesting that Sfrp2 acts as a Wnt agonist. We demonstrate that Sfrp2 plays a role in self-renewal of SSCs and in the recruitment and differentiation of adult SSCs during bone healing. SFRP2 is also a useful marker of BMSC/SSC multipotency, and a factor to potentially improve the quality of ex vivo expanded BMSC/SSC products.
- Published
- 2021
8. Quantitative Craniofacial Analysis and Generation of Human Induced Pluripotent Stem Cells for Muenke Syndrome: A Case Report
- Author
-
Byron W H Mui, Jeremiah W. Woodcock, Randall K. Merling, Vamsee D. Myneni, Kulsum Iqbal, Fahad Kidwai, Pamela Orzechowski, Barbara S. Mallon, Jeffrey W. Gilman, Deepika Arora, Pamela Gehron Robey, Konstantinia Almpani, Paul Kruszka, Maximilian Muenke, Moaz Ahmad, Janice S. Lee, Sriram S Paravastu, Cyrus Keyvanfar, and Priyam Jani
- Subjects
Proband ,business.industry ,Craniofacial abnormality ,QH301-705.5 ,geometric morphometric analysis ,Macrocephaly ,Case Report ,Cell Biology ,Fibroblast growth factor receptor 3 ,Muenke syndrome ,medicine.disease ,Bioinformatics ,Penetrance ,craniofacial abnormalities ,Craniosynostosis ,human induced pluripotent stem cells ,craniosynostosis ,medicine ,medicine.symptom ,Craniofacial ,Biology (General) ,business ,Molecular Biology ,Developmental Biology - Abstract
In this case report, we focus on Muenke syndrome (MS), a disease caused by the p.Pro250Arg variant in fibroblast growth factor receptor 3 (FGFR3) and characterized by uni- or bilateral coronal suture synostosis, macrocephaly without craniosynostosis, dysmorphic craniofacial features, and dental malocclusion. The clinical findings of MS are further complicated by variable expression of phenotypic traits and incomplete penetrance. As such, unraveling the mechanisms behind MS will require a comprehensive and systematic way of phenotyping patients to precisely identify the impact of the mutation variant on craniofacial development. To establish this framework, we quantitatively delineated the craniofacial phenotype of an individual with MS and compared this to his unaffected parents using three-dimensional cephalometric analysis of cone beam computed tomography scans and geometric morphometric analysis, in addition to an extensive clinical evaluation. Secondly, given the utility of human induced pluripotent stem cells (hiPSCs) as a patient-specific investigative tool, we also generated the first hiPSCs derived from a family trio, the proband and his unaffected parents as controls, with detailed characterization of all cell lines. This report provides a starting point for evaluating the mechanistic underpinning of the craniofacial development in MS with the goal of linking specific clinical manifestations to molecular insights gained from hiPSC-based disease modeling.
- Published
- 2021
9. In Vivo Formation of Stable Hyaline Cartilage by Naïve Human Bone Marrow Stromal Cells with Modified Fibrin Microbeads
- Author
-
Sergei A. Kuznetsov, Raphael Gorodetsky, Natasha Cherman, Luis F de Castro, Astar Hailu-Lazmi, and Pamela Gehron Robey
- Subjects
0301 basic medicine ,Stromal cell ,Transplantation, Heterologous ,Mesenchymal Stem Cell Transplantation ,In vivo implantation ,Immunocompromised Host ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,stomatognathic system ,Tissue engineering ,Tissue Engineering and Regenerative Medicine ,Hyaluronic acid ,medicine ,Animals ,Humans ,lcsh:QH573-671 ,Hyaluronic Acid ,lcsh:R5-920 ,Fibrin ,Tissue Engineering ,Tissue Scaffolds ,lcsh:Cytology ,Chemistry ,Hyaline cartilage ,Cartilage ,Bone marrow stromal cells ,Mesenchymal stem cell ,Cell Differentiation ,Mesenchymal Stem Cells ,Cartilage formation ,Cell Biology ,General Medicine ,Microspheres ,3. Good health ,Cell biology ,Hyaline Cartilage ,030104 developmental biology ,medicine.anatomical_structure ,Fibrin microbeads ,Bone marrow ,Stem cell ,lcsh:Medicine (General) ,Chondrogenesis ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Osteoarthritic and other types of articular cartilage defects never heal on their own. Medicinal and surgical approaches are often ineffective, and the supply of autologous chondrocytes for tissue engineering is very limited. Bone marrow stromal cells (BMSCs, also known as bone marrow-derived mesenchymal stem cells) have been suggested as an adequate cell source for cartilage reconstruction. However, the majority of studies employing BMSCs for cartilage tissue engineering have used BMSCs predifferentiated into cartilage prior to implantation. This strategy has failed to achieve formation of stable, hyaline-like cartilage, resistant to hypertrophy in vivo. We hypothesized that in vitro predifferentiation of BMSCs is not necessary when cells are combined with an adequate scaffold that supports the formation of stable cartilage in vivo. In this study, naïve (undifferentiated) human BMSCs were attached to dehydrothermally crosslinked stable fibrin microbeads (FMBs) without and with other scaffolds and implanted subcutaneously into immunocompromised mice. Optimal formation of abundant, hypertrophy-resistant, ectopic hyaline-like cartilage was achieved when BMSCs were attached to FMBs covalently coated with hyaluronic acid. The cartilage that was formed was of human origin and was stable for at least 28 weeks in vivo. Stem Cells Translational Medicine 2019;8:586–592
- Published
- 2019
- Full Text
- View/download PDF
10. Self-organized yolk sac-like organoids allow for scalable generation of multipotent hematopoietic progenitor cells from human induced pluripotent stem cells
- Author
-
Yuting Huang, Isonaka R, Takuya Maeda, Naritaka Tamaoki, Masaki Kimura, Suman K. Vodnala, Naoya U, Pamela Gehron Robey, Sweeney Cl, Choi U, Harry L. Malech, Stroncek Df, Jizhong Zou, Michael J. Kruhlak, Takanori Takebe, Brault J, Goldstein Ds, John F. Tisdale, Raul Vizcardo, Meghan L. Good, Nicholas P. Restifo, Mora Jj, Stefan Siebert, Ha N, Yasuhiro Yamazaki, and Koontz S
- Subjects
food.ingredient ,Stromal cell ,Embryo ,Biology ,Embryonic stem cell ,In vitro ,Cell biology ,food ,medicine.anatomical_structure ,Yolk ,embryonic structures ,Organoid ,medicine ,Functional ability ,Yolk sac - Abstract
SUMMARYThe human definitive yolk sac is an important organ supporting the early developing embryo through nutrient supply and by facilitating the establishment of the embryonic circulatory system. However, the molecular and cellular biology of the human yolk sac remains largely obscure due to the lack of suitable in vitro models. Here, we show that human induced pluripotent stem cells (hiPSCs) co-cultured with various types of stromal cells as spheroids self-organize into yolk sac-like organoids without the addition of exogenous factors. Yolk sac-like organoids recapitulated a yolk sac specific cellular complement and structures as well as the functional ability to generate definitive hematopoietic progenitor cells (HPCs). Furthermore, sequential hemato-vascular ontogenesis could be observed during organoid formation. Notably, our organoid system can be performed in a scalable, autologous, and xeno-free condition, thereby providing an important model of human definitive yolk sac development and allows for efficient bulk generation of hiPSC-derived HPCs.
- Published
- 2021
- Full Text
- View/download PDF
11. Bone Marrow Stromal Cell Assays: In Vitro and In Vivo
- Author
-
Mara Riminucci, Pamela Gehron Robey, Sergei A. Kuznetsov, and Paolo Bianco
- Subjects
endocrine system ,Stromal cell ,Cellular differentiation ,Cell Culture Techniques ,Clinical uses of mesenchymal stem cells ,Stroma ,Biology ,Article ,Colony-Forming Units Assay ,In vivo transplantation ,Mice ,stomatognathic system ,Colony forming unit-fibroblast ,medicine ,Animals ,Humans ,Bone marrow ,Progenitor cell ,Bone ,Bone marrow , Colony-forming unit fi broblast , Bone , Cartilage , Stroma , Marrow adipocytes , In vitro assays , In vivo transplantation ,Gene Expression Profiling ,Mesenchymal stem cell ,Cell Differentiation ,Mesenchymal Stem Cells ,Flow Cytometry ,Cartilage ,In vitro assays ,Marrow adipocytes ,Cell biology ,medicine.anatomical_structure ,Colony-forming unit fi broblast ,Cell culture ,Immunology ,Stem cell - Abstract
Populations of bone marrow stromal cells (BMSCs, also known as bone marrow-derived "mesenchymal stem cells") contain a subset of cells that are able to recapitulate the formation of a bone/marrow organ (skeletal stem cells, SSCs). It is now apparent that cells with similar but not identical properties can be isolated from other skeletal compartments (growth plate, periosteum). The biological properties of BMSCs, and these related stem/progenitor cells, are assessed by a variety of assays, both in vitro and in vivo. Application of these assays in an appropriate fashion provide a great deal of information on the role of BMSCs, and the subset of SSCs, in health and in disease.
- Published
- 2021
12. From stem cells to bone-forming cells
- Author
-
Marta Serafini, Samantha Donsante, Alessandro Corsi, Mara Riminucci, Biagio Palmisano, and Pamela Gehron Robey
- Subjects
Lineage (genetic) ,Cell ,Review ,Biology ,bone ,Bone and Bones ,Catalysis ,Epigenesis, Genetic ,lcsh:Chemistry ,Inorganic Chemistry ,bone marrow stromal cells ,osteoblasts ,skeletal biology ,skeletal stem cells ,Osteogenesis ,medicine ,Animals ,Humans ,Physical and Theoretical Chemistry ,lcsh:QH301-705.5 ,Molecular Biology ,Spectroscopy ,Stem Cells ,Regeneration (biology) ,Organic Chemistry ,Embryonic Stage ,General Medicine ,Embryonic stem cell ,Computer Science Applications ,Cell biology ,medicine.anatomical_structure ,lcsh:Biology (General) ,lcsh:QD1-999 ,Bone forming ,Stem cell ,Function (biology) ,Signal Transduction - Abstract
Bone formation starts near the end of the embryonic stage of development and continues throughout life during bone modeling and growth, remodeling, and when needed, regeneration. Bone-forming cells, traditionally termed osteoblasts, produce, assemble, and control the mineralization of the type I collagen-enriched bone matrix while participating in the regulation of other cell processes, such as osteoclastogenesis, and metabolic activities, such as phosphate homeostasis. Osteoblasts are generated by different cohorts of skeletal stem cells that arise from different embryonic specifications, which operate in the pre-natal and/or adult skeleton under the control of multiple regulators. In this review, we briefly define the cellular identity and function of osteoblasts and discuss the main populations of osteoprogenitor cells identified to date. We also provide examples of long-known and recently recognized regulatory pathways and mechanisms involved in the specification of the osteogenic lineage, as assessed by studies on mice models and human genetic skeletal diseases.
- Published
- 2021
13. Modeling plasticity and dysplasia of pancreatic ductal organoids derived from human pluripotent stem cells
- Author
-
Zahra Dantes, Maximilian Reichert, József Maléth, Thomas Engleitner, Thomas Seufferlein, MK Melzer, Christian M. Cohrs, J Merkle, Martin Wagner, Martin Müller, Tamara Madácsy, Joscha Griger, Cagatay Günes, Stefan Liebau, Markus Breunig, Matthias Meier, Thomas F. E. Barth, Sandra Wiedenmann, Patrick C. Hermann, Stephan Speier, Bernhard Kuster, Sandra Heller, Maximilian Schmid, Gaurav Jain, Pamela Gehron Robey, Johannes Krumm, Florian Kuhn, Lukas Perkhofer, Christian Bolenz, Oliver Wessely, Alexander Kleger, Bence Sipos, Árpád Varga, Ninel Azoitei, Roland Rad, Jana Krüger, and Meike Hohwieler
- Subjects
Pluripotent Stem Cells ,Proteomics ,03. Orvos- és egészségtudomány ,medicine.disease_cause ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,CDKN2A ,Genetics ,GNAS complex locus ,medicine ,Animals ,Humans ,Induced pluripotent stem cell ,030304 developmental biology ,0303 health sciences ,biology ,Pancreatic Ducts ,01.06. Biológiai tudományok ,Cell Biology ,medicine.disease ,digestive system diseases ,3. Good health ,Transplantation ,Organoids ,Pancreatic Neoplasms ,medicine.anatomical_structure ,Dysplasia ,Cdkn2a ,Gnas ,Ipmn ,Kras ,Pdac ,Disease Modelling ,Ductal Pancreatic Organoids ,Human Pluripotent Stem Cells ,In Vitro Differentiation ,Xenograft ,Mutation ,Cancer research ,biology.protein ,Molecular Medicine ,KRAS ,Carcinogenesis ,Pancreas ,030217 neurology & neurosurgery ,Carcinoma, Pancreatic Ductal - Abstract
Personalized invitro models for dysplasia and carcinogenesis in the pancreas have been constrained by insufficient differentiation of human pluripotent stem cells (hPSCs) into the exocrine pancreatic lineage. Here, we differentiate hPSCs into pancreatic duct-like organoids (PDLOs) with morphological, transcriptional, proteomic, and functional characteristics of human pancreatic ducts, further maturing upon transplantation into mice. PDLOs are generated from hPSCs inducibly expressing oncogenic GNAS, KRAS, or KRAS with genetic covariance of lost CDKN2A and from induced hPSCs derived from a McCune-Albright patient. Each oncogene causes a specific growth, structural, and molecular phenotype invitro. While transplanted PDLOs with oncogenic KRAS alone form heterogenous dysplastic lesions or cancer, KRAS with CDKN2A loss develop dedifferentiated pancreatic ductal adenocarcinomas. In contrast, transplanted PDLOs with mutant GNAS lead to intraductal papillary mucinous neoplasia-like structures. Conclusively, PDLOs enable invitro and invivo studies of pancreatic plasticity, dysplasia, and cancer formation from a genetically defined background.
- Published
- 2021
- Full Text
- View/download PDF
14. List of contributors
- Author
-
Bo Abrahamsen, Robert A. Adler, Sara Ajjour, Mohammad Mehdi Alemi, Dennis E. Anderson, Timothy R. Arnett, Mariam A. Assaad, Ghada T. Ballane, Roland Baron, J.H. Duncan Bassett, Douglas C. Bauer, William A. Bauman, Kristen M. Beavers, Sarah D. Berry, John P. Bilezikian, Emmanuel Biver, Dana Bliuc, Lynda F. Bonewald, Adele L. Boskey, Mary L. Bouxsein, Nathalie Bravenboer, Todd T. Brown, Susan V. Bukata, Katelyn Burkhart, Ernesto Canalis, Christopher Cardozo, Alesha B. Castillo, Jane A. Cauley, Jacqueline R. Center, Julia C. Chen, Roberto Civitelli, Adi Cohen, Felicia Cosman, Carolyn J. Crandall, Brooke M. Crawford, Natalie E. Cusano, Francisco J.A. de Paula, Kim Delbaere, David W. Dempster, Dima L. Diab, Ingrid Dick-de-Paula, Linda A. DiMeglio, Matthew T. Drake, Alanna M.K. Dubrovsky, Luca D’Onofrio, Richard Eastell, Grahame J. Elder, Ghada A. El-Hajj Fuleihan, Kristine E. Ensrud, Serge Ferrari, Bernard Freudenthal, Harry K. Genant, Louis C. Gerstenfeld, Lora Giangregorio, Evelien Gielen, Deborah T. Gold, Steven R. Goldring, Catherine M. Gordon, Francesca Gori, Gail A. Greendale, James F. Griffith, Peyman Hadji, Christopher J. Hernandez, Jonathan Hoggatt, Denise K. Houston, Amira I. Hussein, Christopher R. Jacobs, Xuezhi Jiang, James D. Johnston, Risa Kagan, Lamya Karim, Carrie Karvonen-Gutierrez, Wendy B. Katzman, Masanobu Kawai, Sundeep Khosla, Douglas P. Kiel, Saija A. Kontulainen, Paul Kostenuik, Alexandra Krez, Henry Kronenberg, Rajiv Kumar, Nancy E. Lane, Lisa Langsetmo, Michaël R. Laurent, L. Lawenius, Sergey Leikin, William D. Leslie, E. Michael Lewiecki, Minghao Liu, Yi Liu, Stephen R. Lord, Joseph Lorenzo, Nina S. Ma, Naim M. Maalouf, Robert Marcus, Michael R. McClung, Marcela Moraes Mendes, Paul D. Miller, Madhusmita Misra, Mahshid Mohseni, Elise F. Morgan, Suzanne N. Morin, Mona Al Mukaddam, Chris J.J. Mulder, Nandini Nair, Nicola Napoli, Nat Nasomyont, Dorothy A. Nelson, Jeri W. Nieves, Robert Nissenson, Claes Ohlsson, Christina V. Oleson, Laura Ortinau, Eric Orwoll, Susan M. Ott, Roberto Pacifici, Andrea Palermo, A.M. Parfitt, Dongsu Park, Sylvain Provot, Sonia Bhandari Randhawa, John F. Randolph, Fernando Rivadeneira, Pamela Gehron Robey, Lauren Robinson, Tara Rogers-Soeder, G. David Roodman, Clifford J. Rosen, Kenneth G. Saag, Shivani Sahni, Khashayar Sakhaee, David T. Scadden, Anne L. Schafer, Ernestina Schipani, Monica C. Serra, Jay R. Shapiro, Catherine Sherrington, James M. Shikany, Shonni J. Silverberg, Andrea J. Singer, K. Sjögren, Peter J. Snyder, Emily M. Stein, Christine M. Swanson, Pawel Szulc, Pamela Taxel, Peter J. Tebben, Sarah E. Twardowski, André G. Uitterlinden, Rachana Vaidya, Cristianna Vallera, Adriaan A. van Bodegraven, Bram C.J. van der Eerden, Marjolein C.H. van der Meulen, André J. van Wijnen, Dirk Vanderschueren, Jean Wactawski-Wende, Laura Watts, Nelson B. Watts, Ashley A. Weaver, Robert S. Weinstein, Graham R. Williams, Joy Wu, Karin C. Wu, Michael T. Yin, Elaine W. Yu, and Hua Zhou
- Published
- 2021
- Full Text
- View/download PDF
15. A single day of TGF-β1 exposure activates chondrogenic and hypertrophic differentiation pathways in bone marrow-derived stromal cells
- Author
-
Pamela Gehron Robey, Travis J. Klein, Michael R. Doran, Kathryn Futrega, and Ross Crawford
- Subjects
0301 basic medicine ,Cartilage, Articular ,Stromal cell ,QH301-705.5 ,Cellular differentiation ,Medicine (miscellaneous) ,Stem-cell differentiation ,Bone Marrow Cells ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Transforming Growth Factor beta1 ,03 medical and health sciences ,0302 clinical medicine ,Chondrocytes ,medicine ,Humans ,Tissue formation ,Biology (General) ,Tissue Engineering ,Sequence Analysis, RNA ,Mesenchymal stem cell ,Hypertrophy ,Chondrogenesis ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Musculoskeletal models ,Mesenchymal stem cells ,Bone marrow ,General Agricultural and Biological Sciences ,030217 neurology & neurosurgery ,Transforming growth factor - Abstract
Virtually all bone marrow-derived stromal cell (BMSC) chondrogenic induction cultures include greater than 2 weeks exposure to transforming growth factor-β (TGF-β), but fail to generate cartilage-like tissue suitable for joint repair. Herein we used a micro-pellet model (5 × 103 BMSC each) to determine the duration of TGF-β1 exposure required to initiate differentiation machinery, and to characterize the role of intrinsic programming. We found that a single day of TGF-β1 exposure was sufficient to trigger BMSC chondrogenic differentiation and tissue formation, similar to 21 days of TGF-β1 exposure. Despite cessation of TGF-β1 exposure following 24 hours, intrinsic programming mediated further chondrogenic and hypertrophic BMSC differentiation. These important behaviors are obfuscated by diffusion gradients and heterogeneity in commonly used macro-pellet models (2 × 105 BMSC each). Use of more homogenous micro-pellet models will enable identification of the critical differentiation cues required, likely in the first 24-hours, to generate high quality cartilage-like tissue from BMSC., Futrega et al. challenge the long established chondrogenic differentiation protocol used to induce bone marrow-derived stromal cell (BMSC) differentiation which presumes the need for more than 2 weeks of continuous TGF-β1 exposure. Using a micro-pellet model, they show that BMSC differentiation is triggered by a single day of TGF-β1 exposure.
- Published
- 2021
16. Activated Gs⍺ pathway and estrogens reveal different subsets of adiponectin-expressing osteoprogenitors within bone marrow stroma
- Author
-
Biagio Palmisano, Rossella Labella, Samantha Donsante, Ilenia Coletta, Giorgia Farinacci, Michele Dello Spedale Venti, Pamela Gehron Robey, Alessandro Corsi, and Mara Riminucci
- Subjects
Endocrinology, Diabetes and Metabolism ,Orthopedics and Sports Medicine - Published
- 2022
- Full Text
- View/download PDF
17. Remembering Dr John D Termine
- Author
-
Ernesto Canalis, T. John Martin, Pamela Gehron Robey, and Marian F. Young
- Subjects
History ,Endocrinology, Diabetes and Metabolism ,MEDLINE ,Library science ,Orthopedics and Sports Medicine - Published
- 2021
- Full Text
- View/download PDF
18. Cell-Mediated Calcification In Vitro
- Author
-
Pamela Gehron Robey
- Subjects
Chemistry ,medicine ,medicine.disease ,Cell mediated immunity ,In vitro ,Cell biology ,Calcification - Published
- 2020
- Full Text
- View/download PDF
19. Characterisation of ovine bone marrow-derived stromal cells (oBMSC) and evaluation of chondrogenically induced micro-pellets for cartilage tissue repair in vivo
- Author
-
Michael R. Doran, Kathryn Futrega, Travis J. Klein, Stan Gronthos, Ena Music, Ross Crawford, Pamela Gehron Robey, and Siamak Saifzadeh
- Subjects
chemistry.chemical_compound ,medicine.anatomical_structure ,Stromal cell ,IBMX ,chemistry ,In vivo ,Adipogenesis ,Cartilage ,medicine ,CD146 ,Bone marrow ,Chondrogenesis ,Cell biology - Abstract
BackgroundBone marrow stromal cells (BMSC) show promise in cartilage repair, and sheep are the most common large animal pre-clinical model. The objective of this study was to characterize ovine BMSC (oBMSC) in vitro, and to evaluate the capacity of chondrogenic micro-pellets manufactured from oBMSC or ovine articular chondrocytes (oACh) to repair osteochondral defects in sheep.MethodsoBMSC were characterised for surface marker expression using flow cytometry and evaluated for tri-lineage differentiation. oBMSC micro-pellets were manufactured in a microwell platform, and chondrogenesis was compared at 2%, 5%, and 20% O2. The capacity of cartilage micro-pellets manufactured from oBMSC or oACh to repair osteochondral defects in adult sheep was evaluated in an 8-week pilot study. Expanded oBMSC were positive for CD44 and CD146 and negative for CD45.ResultsThe common adipogenic induction medium ingredient, 3-Isobutyl-1-methylxanthine (IBMX) was toxic to oBMSC, but adipogenesis could be restored by excluding IBMX from the medium. BMSC chondrogenesis was optimal in a 2% O2 atmosphere. Micro-pellets formed from oBMSC or oACh appeared morphologically similar, but hypertrophic genes were elevated in oBMSC micro-pellets. While oACh micro-pellets formed cartilage-like repair tissue in sheep, oBMSC micro-pellets did not.ConclusionThe sensitivity of oBMSC to IBMX highlights species-species differences between oBMSC and hBMSC. Micro-pellets manufactured from oBMSC were not effective in repairing osteochondral defects, while oACh micro-pellets enabled modest repair. While oBMSC can be driven to form cartilage-like tissue in vitro, their effective use in cartilage repair will require mitigation of hypertrophy.
- Published
- 2020
- Full Text
- View/download PDF
20. Generation of human induced pluripotent stem cell line (NIDCRi001-A) from a Muenke syndrome patient with an FGFR3 p.Pro250Arg mutation
- Author
-
Barbara S. Mallon, Paul Kruszka, Pamela Gehron Robey, Janice S. Lee, Ariel F. Martinez, Fahad Kidwai, Byron W H Mui, Deepika Arora, and Maximilian Muenke
- Subjects
0301 basic medicine ,Induced Pluripotent Stem Cells ,Biology ,medicine.disease_cause ,Article ,Muenke syndrome ,Craniosynostosis ,03 medical and health sciences ,Craniosynostoses ,0302 clinical medicine ,medicine ,Humans ,Receptor, Fibroblast Growth Factor, Type 3 ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,Mutation ,Cell Biology ,General Medicine ,medicine.disease ,Phenotype ,030104 developmental biology ,lcsh:Biology (General) ,Cancer research ,Mutation testing ,Teratoma ,Reprogramming ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Muenke syndrome is the leading genetic cause of craniosynostosis and results in a variety of disabling clinical phenotypes. To model the disease and study the pathogenic mechanisms, a human induced pluripotent stem cell (hiPSC) line was generated from a patient diagnosed with Muenke syndrome. Successful reprogramming was validated by morphological features, karyotyping, loss of reprogramming factors, expression of pluripotency markers, mutation analysis and teratoma formation.
- Published
- 2020
21. List of Contributors
- Author
-
David Abraham, Maria Almeida, Elena Ambrogini, Andrew Arnold, Bence Bakos, Clemens Bergwitz, Daniel D. Bikle, John P. Bilezikian, Neil Binkley, Alessandro Bisello, L.F. Bonewald, George Bou-Gharios, Roger Bouillon, Mary L. Bouxsein, Brendan F. Boyce, Steven Boyd, Maria Luisa Brandi, David B. Burr, Laura M. Calvi, Ernesto Canalis, Xu Cao, Geert Carmeliet, Thomas O. Carpenter, Wenhan Chang, Shek Man Chim, Shilpa Choudhary, Sylvia Christakos, Yong-Hee Patricia Chun, Cristiana Cipriani, Roberto Civitelli, Thomas L. Clemens, Michael T. Collins, Caterina Conte, Mark S. Cooper, Jillian Cornish, Serge Cremers, Bess Dawson-Hughes, Benoit de Crombrugghe, Hector F. DeLuca, David W. Dempster, Matthew T. Drake, Patricia Ducy, Frank H. Ebetino, Klaus Engelke, Reinhold G. Erben, David R. Eyre, Charles R. Farber, Marina Feigenson, Mathieu Ferron, Pablo Florenzano, Francesca Fontana, Brian L. Foster, Peter A. Friedman, Seiji Fukumoto, Laura W. Gamer, Thomas J. Gardella, Patrick Garnero, Harry K. Genant, Francesca Giusti, Andy Göbel, David Goltzman, Jeffrey P. Gorski, James Griffith, R. Graham G Russell, Kurt D. Hankenson, Fadil M. Hannan, Stephen E. Harris, Iris R. Hartley, Christine Hartmann, Robert P. Heaney, Geoffrey N. Hendy, Matthew J. Hilton, Lorenz C. Hofbauer, Gill Holdsworth, Yi-Hsiang Hsu, David M. Hudson, Marja Hurley, Karl L. Insogna, Robert L. Jilka, Mark L. Johnson, Rachelle W. Johnson, Glenville Jones, Stefan Judex, Harald Jüppner, Ivo Kalajzic, Gérard Karsenty, Hua Zhu Ke, Sundeep Khosla, Douglas P. Kiel, J. Klein-Nulend, Frank C. Ko, Yasuhiro Kobayashi, Martin Konrad, Paul J. Kostenuik, Christopher S. Kovacs, Richard Kremer, Venkatesh Krishnan, Henry M. Kronenberg, Peter A. Lakatos, Uri A. Liberman, Joseph A. Lorenzo, Conor C. Lynch, Karen M. Lyons, Y. Linda Ma, Christa Maes, Michael Mannstadt, Stavros Manolagas, Robert Marcus, David E. Maridas, Pierre J. Marie, Francesca Marini, Jasna Markovac, T. John Martin, Brya G. Matthews, Antonio Maurizi, Sasan Mirfakhraee, Sharon M. Moe, David G. Monroe, Carolina A. Moreira, Ralph Müller, David S. Musson, Teruyo Nakatani, Dorit Naot, Nicola Napoli, Tally Naveh-Many, Edward F. Nemeth, Thomas L. Nickolas, Michael S. Ominsky, Noriaki Ono, David M. Ornitz, Nicola C. Partridge, Vihitaben S. Patel, J. Wesley Pike, Carol Pilbeam, Lori Plum, John T. Potts, J. Edward Puzas, Tilman D. Rachner, Audrey Rakian, Rubie Rakian, Nora E. Renthal, Julie A. Rhoades (Sterling), Mara Riminucci, Scott J. Roberts, Pamela Gehron Robey, Michael J. Rogers, G. David Roodman, Clifford J. Rosen, Vicki Rosen, David W. Rowe, Janet Rubin, Clinton T. Rubin, Karl P. Schlingmann, Ego Seeman, Markus J. Seibel, Chris Sempos, Dolores M. Shoback, Caroline Silve, Justin Silver, Natalie A. Sims, Frederick R. Singer, Joseph P. Stains, Steve Stegen, Paula H. Stern, Gaia Tabacco, Istvan Takacs, Naoyuki Takahashi, Donovan Tay, Anna Teti, Rajesh V. Thakker, Ryan E. Tomlinson, Francesco Tonelli, Dwight A. Towler, Elena Tsourdi, Chia-Ling Tu, Nobuyuki Udagawa, Connie M. Weaver, Marc N. Wein, Lee S. Weinstein, MaryAnn Weis, Michael P. Whyte, Bart O. Williams, Xin Xu, Shoshana Yakar, Yingzi Yang, Stefano Zanotti, and Hong Zhou
- Published
- 2020
- Full Text
- View/download PDF
22. Skeletal stem cells
- Author
-
Mara Riminucci and Pamela Gehron Robey
- Subjects
Haematopoiesis ,medicine.anatomical_structure ,Stromal cell ,Stroma ,Cartilage ,medicine ,Bone marrow ,Stem cell ,Progenitor cell ,Biology ,Bone resorption ,Cell biology - Abstract
Skeletal stem cells (SSCs) are a subset of bone marrow stromal cells (BMSCs), found as pericytes on the abluminal side of marrow sinusoids. These cells have the remarkable feature of forming cartilage, bone, stroma that supports blood formation, and marrow adipocytes, based on rigorous differentiation assays. They also have the ability to self-renew, making them bona fide postnatal stem cells. These cells are essential not only for continuous bone formation, but also in the control of bone resorption based on their support of hematopoietic cells and control of osteoclastic precursors. Consequently, these cells are central mediators of bone homeostasis. As such, changes in their biological activity due to mutation (intrinsic) or due to changes in the microenvironment (extrinsic) will result in a skeletal disease or disorder, and sometimes disruption of hematopoiesis. Last, SSCs/BMSCs are a promising source of cells for rebuilding bone lost to trauma, surgery, or disease.
- Published
- 2020
- Full Text
- View/download PDF
23. Changes in gene expression in human skeletal stem cells transduced with constitutively active Gsα correlates with hallmark histopathological changes seen in fibrous dysplastic bone
- Author
-
Fiammetta Vernì, Domenico Raimondo, Agnese Persichetti, Alessandro Corsi, Enrico Tagliafico, Mara Riminucci, Romina Burla, Isabella Saggio, Mattia La Torre, Simona Del Giudice, Letizia Astrologo, Giuseppe Giannicola, Cristina Remoli, Pamela Gehron Robey, School of Biological Sciences, and NTU Institute of Structural Biology
- Subjects
0301 basic medicine ,Receptor complex ,Physiology ,Gene Expression ,Datasets as Topic ,Pathology and Laboratory Medicine ,Biochemistry ,Fats ,0302 clinical medicine ,Animal Cells ,Osteogenesis ,Medicine and Health Sciences ,Adipocytes ,GTP-Binding Protein alpha Subunits, Gs ,macroarrarry ,fibrous dysplasia ,GNAS ,R201C ,Cells, Cultured ,Connective Tissue Cells ,Oligonucleotide Array Sequence Analysis ,Multidisciplinary ,Adipogenesis ,Gene Ontologies ,Stem Cells ,Biological sciences [Science] ,Cell Differentiation ,Genomics ,Lipids ,Healthy Volunteers ,Recombinant Proteins ,Cell biology ,Up-Regulation ,Haematopoiesis ,Connective Tissue ,030220 oncology & carcinogenesis ,Gain of Function Mutation ,Medicine ,Signal transduction ,Stem cell ,Cellular Types ,Anatomy ,Research Article ,Dysplasia ,Science ,Adipose Tissue, White ,Primary Cell Culture ,Bone Marrow Cells ,Biology ,03 medical and health sciences ,Signs and Symptoms ,Downregulation and upregulation ,Diagnostic Medicine ,GNAS complex locus ,Genetics ,Chromogranins ,Humans ,Computer Simulation ,Progenitor cell ,Osteoblasts ,Cyclic Nucleotide Phosphodiesterases, Type 7 ,Gene Expression Profiling ,Biology and Life Sciences ,Computational Biology ,Cell Biology ,Fibrous Dysplasia of Bone ,Genome Analysis ,Hematopoiesis ,ADAM Proteins ,030104 developmental biology ,Biological Tissue ,Cell culture ,biology.protein ,CCAAT-Enhancer-Binding Proteins ,Stromal Cells ,Physiological Processes - Abstract
Fibrous dysplasia (FD) of bone is a complex disease of the skeleton caused by dominant activating mutations of the GNAS locus encoding for the α subunit of the G protein-coupled receptor complex (Gsα). The mutation involves a substitution of arginine at position 201 by histidine or cysteine (GsαR201H or R201C), which leads to overproduction of cAMP. Several signaling pathways are implicated downstream of excess cAMP in the manifestation of disease. However, the pathogenesis of FD remains largely unknown. The overall FD phenotype can be attributed to alterations of skeletal stem/progenitor cells which normally develop into osteogenic or adipogenic cells (in cis), and are also known to provide support to angiogenesis, hematopoiesis, and osteoclastogenesis (in trans). In order to dissect the molecular pathways rooted in skeletal stem/progenitor cells by FD mutations, we engineered human skeletal stem/progenitor cells with the GsαR201C mutation and performed transcriptomic analysis. Our data suggest that this FD mutation profoundly alters the properties of skeletal stem/progenitor cells by pushing them towards formation of disorganized bone with a concomitant alteration of adipogenic differentiation. In addition, the mutation creates an altered in trans environment that induces neovascularization, cytokine/chemokine changes and osteoclastogenesis. In silico comparison of our data with the signature of FD craniofacial samples highlighted common traits, such as the upregulation of ADAM (A Disintegrin and Metalloprotease) proteins and other matrix-related factors, and of PDE7B (Phosphodiesterase 7B), which can be considered as a buffering process, activated to compensate for excess cAMP. We also observed high levels of CEBPs (CCAAT-Enhancer Binding Proteins) in both data sets, factors related to browning of white fat. This is the first analysis of the reaction of human skeletal stem/progenitor cells to the introduction of the FD mutation and we believe it provides a useful background for further studies on the molecular basis of the disease and for the identification of novel potential therapeutic targets. Published version
- Published
- 2020
24. Bone Marrow Stromal Cells Assays-In Vitro and In Vivo
- Author
-
Pamela Gehron Robey, Kuznetsov, Sergei A., Bianco, Paolo, and Riminucci, Mara
- Subjects
bone marrow ,in vivo transplantation ,stroma ,marrow adipocytes ,in vitro assays ,bone marrow, colony forming unit-fibroblast, bone, cartilage, stroma, marrow adipocytes, in vitro assays, in vivo transplantation ,cartilage ,bone ,colony forming unit-fibroblast - Published
- 2020
25. Combinatorial cassettes to systematically evaluate tissue-engineered constructs in recipient mice
- Author
-
Carl G. Simon, Maeda Azusa, Pamela Gehron Robey, Subhadip Bodhak, Luis F de Castro, Danielle Bonfim, and Sergei A. Kuznetsov
- Subjects
Calcium Phosphates ,0301 basic medicine ,Stromal cell ,Biophysics ,Bone Morphogenetic Protein 2 ,Bioengineering ,Biology ,Bone morphogenetic protein 2 ,Article ,Bone tissue engineering ,Biomaterials ,Mice ,03 medical and health sciences ,Tissue engineering ,Osteogenesis ,In vivo ,Gene expression ,Animals ,Humans ,Polytetrafluoroethylene ,Cells, Cultured ,Tissue Engineering ,Tissue Scaffolds ,Mesenchymal Stem Cells ,Histology ,Durapatite ,030104 developmental biology ,Real-time polymerase chain reaction ,Mechanics of Materials ,Bone Substitutes ,Ceramics and Composites ,Gelatin ,Female ,Porosity ,Biomedical engineering - Abstract
Ectopic bone formation in mice is the gold standard for evaluation of osteogenic constructs. By regular procedures, usually only 4 constructs can be accommodated per mouse, limiting screening power. Combinatorial cassettes (combi-cassettes) hold up to 19 small, uniform constructs from the time of surgery, through time in vivo, and subsequent evaluation. Two types of bone tissue engineering constructs were tested in the combi-cassettes: i) a cell-scaffold construct containing primary human bone marrow stromal cells with hydroxyapatite/tricalcium phosphate particles (hBMSCs + HA/TCP) and ii) a growth factor-scaffold construct containing bone morphogenetic protein 2 in a gelatin sponge (BMP2+GS). Measurements of bone formation by histology, bone formation by X-ray microcomputed tomography (μCT) and gene expression by quantitative polymerase chain reaction (qPCR) showed that constructs in combi-cassettes were similar to those created by regular procedures. Combi-cassettes afford placement of multiple replicates of multiple formulations into the same animal, which enables, for the first time, rigorous statistical assessment of: 1) the variability for a given formulation within an animal (intra-animal variability), 2) differences between different tissue-engineered formulations within the same animal and 3) the variability for a given formulation in different animals (inter-animal variability). Combi-cassettes enable a more high-throughput, systematic approach to in vivo studies of tissue engineering constructs.
- Published
- 2018
- Full Text
- View/download PDF
26. Activation of RANK/RANKL/OPG Pathway Is Involved in the Pathophysiology of Fibrous Dysplasia and Associated With Disease Burden
- Author
-
Jeffrey Y. Tsai, Alison M. Boyce, Andrea Burke, Howard D. Wang, Luis F de Castro, Rachel I Gafni, Michael T. Collins, Pamela Gehron Robey, Kristen S. Pan, Alfredo A. Molinolo, Nisan Bhattacharyya, and Pablo Florenzano
- Subjects
Male ,musculoskeletal diseases ,0301 basic medicine ,medicine.medical_specialty ,Stromal cell ,Endocrinology, Diabetes and Metabolism ,Bone Marrow Cells ,030209 endocrinology & metabolism ,Article ,Bone remodeling ,03 medical and health sciences ,0302 clinical medicine ,Osteoprotegerin ,Internal medicine ,Humans ,Medicine ,Orthopedics and Sports Medicine ,Prostaglandin E2 ,Cells, Cultured ,Receptor Activator of Nuclear Factor-kappa B ,biology ,business.industry ,Monocyte ,RANK Ligand ,Mesenchymal Stem Cells ,Fibrous Dysplasia of Bone ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Denosumab ,RANKL ,biology.protein ,Female ,Bone marrow ,business ,Signal Transduction ,medicine.drug - Abstract
Fibrous dysplasia of bone (FD) is a mosaic disease caused by mutations in GNAS. Constitutive activation of the α-subunit of the Gs stimulatory protein (Gαs) leads to dysregulated proliferation of bone marrow stromal cells (BMSCs), generating expansile lesions of fibrotic tissue and abnormal bone. Local bone remodeling regulation by BMSCs is also altered, and FD tissue is characterized by abundant osteoclast-like cells that may be essential for lesion expansion. Animal models show local expression of RANKL in bone lesions, and treatment with the RANKL neutralizing antibody denosumab decreased lesion expansion rate in a patient with aggressive FD. However, the role of RANKL/osteoprotegerin (OPG) in FD pathophysiology is not yet understood. We measured serum levels of RANKL, OPG, and inactive RANKL-OPG complexes in FD patients of known disease burden and in healthy volunteers (HVs). RANK, RANKL, and Ki67 immunohistochemistry were assessed in FD tissue. Cultured FD and HV BMSCs were stimulated with prostaglandin E2 (PGE2 ) and 1,25 vitamin D3 to increase RANKL expression, and media levels of RANKL and OPG were measured. Osteoclastogenic induction by FD or HV BMSCs was assessed in co-cultures with HV peripheral monocytes. FD patients showed a 16-fold increase in serum RANKL compared to HVs. OPG was moderately increased (24%), although RANKL/OPG ratio was 12-fold higher in FD patients than in HVs. These measurements were positively correlated with the skeletal burden score (SBS), a validated marker of overall FD burden. No differences in serum inactive RANKL-OPG complexes were observed. In FD tissue, RANKL+ and Ki67+ fibroblastic cells were observed near RANK+ osteoclasts. High levels of RANKL were released by FD BMSCs cultures, but were undetectable in HV cultures. FD BMSC released less OPG than HV BMSCs. FD, but not HV BMSCs, induced osteoclastogenesis in monocyte co-cultures, which was prevented by denosumab addition. These data are consistent with the role of RANKL as a driver in FD-induced osteoclastogenesis. © 2018 American Society for Bone and Mineral Research.
- Published
- 2018
- Full Text
- View/download PDF
27. Proceedings of the signature series symposium 'cellular therapies for orthopaedics and musculoskeletal disease proven and unproven therapies—promise, facts and fantasy,' international society for cellular therapies, montreal, canada, may 2, 2018
- Author
-
Frank Barry, Ivan Martin, Thomas W. Bauer, Cecilia Pascual-Garrido, Christian Jorgensen, Scott A. Rodeo, George F. Muschler, Constance R. Chu, Jérôme Guicheux, Pamela Gehron Robey, Nicolas S. Piuzzi, Massimo Dominici, Stéphane Maddens, M. A.R.C. Long, J. O.H.N. Barrett, David Karli, Richard McFarland, Johnny Huard, Laurie R. Goodrich, Daniel J. Weiss, Department of Orthopedic Surgery [Cleveland, Ohio, USA], Cleveland Clinic, Instituto Universitario del Hospital Italiano [Buenos Aires, Argentina], Department of Medical and Surgical Sciences for Children and Adults [Modena, Italy] (Laboratory of Cellular Therapy), Università degli Studi di Modena e Reggio Emilia (UNIMORE), MTF Biologics, Edison [New Jersey, USA], Adult Reconstruction-Adolescent and Young Adult Hip Service [St. Louis, Missouri, USA] (School of Medicine), Washington University in Saint Louis (WUSTL), Orthopaedic Soft Tissue Research Program [New York, NY, USA], Hospital for Special Surgery, Department of Orthopaedic Surgery [Houston, TX, USA], The University of Texas Health Science Center at Houston (UTHealth), Steadman Philippon Research Institute, Regenerative Medicine and Skeleton research lab (RMeS), Ecole Nationale Vétérinaire, Agroalimentaire et de l'alimentation Nantes-Atlantique (ONIRIS)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM), Service d'Oto-Rhino-Laryngologie et de Chirurgie Cervico-Faciale [CHU Nantes] (PHU4 - OTONN), Centre hospitalier universitaire de Nantes (CHU Nantes), Advanced Regenerative Manufacturing Institute [Manchester, NH, USA], Standards Coordinating Body, Department of Clinical Sciences [Fort Collins, CO, USA] (Orthopaedic Research Center), Colorado State University [Fort Collins] (CSU), Vetbiobank [Marcy l’Etoile, France], Department of Health and Human Services [Bethesda, MD, USA] (Skeletal Biology Section ), National Institutes of Health [Bethesda] (NIH), Department of Pathology and Laboratory Medicine [New-York, NY, USA], Stem Cell Allogeneic Transplant Section [Bethesda, MD, USA], Regenerative Medicine Institute [Galway, Ireland], National University of Ireland [Galway] (NUI Galway), Greyledge Technologies - LLC [Vail, CO, USA], Department of Orthopaedic Surgery [Stanford], Stanford Medicine, Stanford University-Stanford University, Veterans Affairs Palo Alto Health Care System [Palo Alto, CA, USA], University of Vermont [Burlington], Department of Biomedicine [Basel], University Hospital Basel [Basel], Unité thérapeutique d'immunologie clinique et des maladies ostéoarticulaires [Hôpital Lapeyronie, Montpellier], Hôpital Lapeyronie [Montpellier] (CHU), The authors thanks both the ISCT and the sponsors of the First Signature Series Symposium 'Cellular Therapies for Orthopaedics and Musculoskeletal Disease Proven and Unproven Therapies–Promise, Facts and Fantasy,' May 2, 2018, Montreal, Canada: Greyledge Technologies (Edwards, Colorado), MTF Biologics (Edison, New Jersey), Orthofix (Lewisville, Texas), MEdXcell (Lausanne, Switzerland), Osiris Therapeutics (Columbia, Maryland) and Angiocrine Bioscience (San Diego, California). Additionally this work was supported, in part, by the DIR, NIDCR, a part of the Intramural Research Program (IRP), NIH, DHHS (to P.G.R., ZIA DE000380)., Università degli Studi di Modena e Reggio Emilia = University of Modena and Reggio Emilia (UNIMORE), Regenerative Medicine and Skeleton (RMeS), École nationale vétérinaire, agroalimentaire et de l'alimentation Nantes-Atlantique (ONIRIS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN), Jehan, Frederic, Ecole Nationale Vétérinaire, Agroalimentaire et de l'alimentation Nantes-Atlantique (ONIRIS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), and University of Vermont College of Medicine [Burlington, VT, USA]
- Subjects
Immunology and Allergy ,Immunology ,Oncology ,Genetics (clinical) ,Cell Biology ,Transplantation ,Cancer Research ,0301 basic medicine ,medicine.medical_specialty ,education ,Article ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Fantasy ,Misinformation ,health care economics and organizations ,Confusion ,[SDV.MHEP.RSOA] Life Sciences [q-bio]/Human health and pathology/Rhumatology and musculoskeletal system ,030222 orthopedics ,Government ,[SDV.MHEP.GEG] Life Sciences [q-bio]/Human health and pathology/Geriatry and gerontology ,business.industry ,[SDV.MHEP.GEG]Life Sciences [q-bio]/Human health and pathology/Geriatry and gerontology ,Musculoskeletal disease ,humanities ,3. Good health ,030104 developmental biology ,[SDV.MHEP.RSOA]Life Sciences [q-bio]/Human health and pathology/Rhumatology and musculoskeletal system ,Orthopedic surgery ,Engineering ethics ,medicine.symptom ,business - Abstract
International audience; The Signature Series Symposium "Cellular Therapies for Orthopaedics and Musculoskeletal Disease Proven and Unproven Therapies-Promise, Facts and Fantasy" was held as a pre-meeting of the 26th International Society for Cellular Therapy (ISCT) annual congress in Montreal, Canada, May 2, 2018. This was the first ISCT program that was entirely dedicated to the advancement of cell-based therapies for musculoskeletal diseases. Cellular therapies in musculoskeletal medicine are a source of great promise and opportunity. They are also the source of public controversy, confusion and misinformation. Patients, clinicians, scientists, industry and government share a commitment to clear communication and responsible development of the field. Therefore, this symposium convened thought leaders from around the world in a forum designed to catalyze communication and collaboration to bring the greatest possible innovation and value to patients with musculoskeletal conditions.
- Published
- 2018
- Full Text
- View/download PDF
28. The Composition of Bone
- Author
-
Pamela Gehron Robey and Adele L. Boskey
- Subjects
Extracellular matrix ,0301 basic medicine ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Chemical engineering ,Chemistry ,030220 oncology & carcinogenesis ,Composition (visual arts) ,Food science - Published
- 2018
- Full Text
- View/download PDF
29. Continuing Challenges in Advancing Preclinical Science in Skeletal Cell-Based Therapies and Tissue Regeneration
- Author
-
Pamela Gehron Robey, David W. Rowe, and Joseph Featherall
- Subjects
0301 basic medicine ,business.industry ,Endocrinology, Diabetes and Metabolism ,Key issues ,law.invention ,Cell therapy ,Data sharing ,03 medical and health sciences ,030104 developmental biology ,Conceptual framework ,law ,CLARITY ,Medicine ,Orthopedics and Sports Medicine ,Engineering ethics ,State of the science ,business ,Cell based - Abstract
Cell-based therapies hold much promise for musculoskeletal medicine; however, this rapidly growing field faces a number of challenges. Few of these therapies have proven clinical benefit, and an insufficient regulatory environment has allowed for widespread clinical implementation without sufficient evidence of efficacy. The technical and biological complexity of cell-based therapies has contributed to difficulties with reproducibility and mechanistic clarity. In order to aid in addressing these challenges, we aim to clarify the key issues in the preclinical cell therapy field, and to provide a conceptual framework for advancing the state of the science. Broadly, these suggestions relate to: (i) delineating cell-therapy types and moving away from "catch-all" terms such as "stem cell" therapies; (ii) clarifying descriptions of cells and their processing; and (iii) increasing the standard of in vivo evaluation of cell-based therapy experiments to determining cell fates. Further, we provide an overview of methods for experimental evaluation, data sharing, and professional society participation that would be instrumental in advancing this field. © 2018 American Society for Bone and Mineral Research.
- Published
- 2018
- Full Text
- View/download PDF
30. Comparison of human bone marrow stromal cells cultured in human platelet growth factors and fetal bovine serum
- Author
-
Steven J. Chen, Ping Jin, Katherine Tran, Jiaqiang Ren, David F. Stroncek, Pamela Gehron Robey, Marianna Sabatino, and Dawn C. Ward
- Subjects
Serum ,0301 basic medicine ,Stromal cell ,medicine.medical_treatment ,Fetal bovine serum ,Cell Culture Techniques ,lcsh:Medicine ,Bone Marrow Cells ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,PEDF ,medicine ,Animals ,Humans ,Platelet lysate ,Cells, Cultured ,Cell Proliferation ,Platelet-Derived Growth Factor ,Chemistry ,Research ,Growth factor ,Mesenchymal stem cell ,Bone marrow stromal cells ,lcsh:R ,General Medicine ,Potency ,equipment and supplies ,Molecular biology ,MicroRNAs ,030104 developmental biology ,Cytokine ,medicine.anatomical_structure ,Gene Expression Regulation ,Cytokines ,Mesenchymal stem cells ,Cattle ,lipids (amino acids, peptides, and proteins) ,Bone marrow ,Lymphocyte Culture Test, Mixed ,Stromal Cells ,Transcriptome ,Biomarkers - Abstract
Background Bone marrow stromal cells (BMSCs) have classically been cultured in media supplemented with fetal bovine serum (FBS). As an alternative to FBS, pooled solvent detergent apheresis platelets, HPGF-C18, was evaluated for BMSC culture. Methods A comparison of passage 2 BMSC growth revealed that 10% HPGF-C18 produced similar cell numbers as 20% FBS. Marrow aspirates from 5 healthy subjects were cultured for 4 passages in 10% HPGF-C18 or 20% FBS and were analyzed for proliferation, colony formation efficiency (CFE), surface marker expression, suppression of mixed lymphocyte reactions (MLRs), global gene and microRNA expression analysis. BMSC supernatant cytokine and growth factor concentrations were also compared. Results Primary cultures of marrow aspirates in 10% HPGF-C18 and 20% FBS yielded similar numbers and CFE. After 4 passages, 10% HPGF-C18 and 20% FBS yielded similar numbers of BMSCs, surface marker expression patterns and immunosuppression effects. Gene and microRNA expression analysis revealed that BMSCs cultured under the two conditions had distinct expression profiles. Gene Set Enrichment Analysis (GSEA) revealed HPGF-C18-cultured BMSCs were enriched in metabolic processing and biosynthetic pathways, cell proliferation and cell cycle pathways, and immune response pathways. FBS-cultured BMSCs were enriched in MAPK signaling, TGF-beta signaling, cell adhesion and extracellular matrix pathways. Differently expressed microRNAs were related to the osteogenesis of BMSCs. The supernatant of HPGF-C18 BMSCs had higher levels of PEDF and TGFB1 and lower levels of IL6, VEGF, SDF1 and PLGF. Conclusions Traditional measures, expansion, surface marker expression and inhibition of MLRs suggest that BMSC cultured in HPGF-C18 and FBS were similar, but analysis at the molecular level revealed many differences. BMSCs cultured in HPGF-C18 should be assessed in specific functional assays that reflect application-specific potency before substituting FBS with HPGF-C18.
- Published
- 2018
- Full Text
- View/download PDF
31. Articular cartilage regeneration in rats using human bone marrow stromal cells
- Author
-
S.J. Gadomski, S. Kuznetsov, S.S. Paravastu, B.W. Mui, R. Merling, and Pamela Gehron Robey
- Subjects
Pathology ,medicine.medical_specialty ,Stromal cell ,Rheumatology ,Chemistry ,Regeneration (biology) ,Biomedical Engineering ,medicine ,Human bone ,Orthopedics and Sports Medicine ,Articular cartilage - Published
- 2021
- Full Text
- View/download PDF
32. Clear up this stem-cell mess
- Author
-
Douglas Sipp, Pamela Gehron Robey, and Leigh Turner
- Subjects
0301 basic medicine ,Multidisciplinary ,business.industry ,Regeneration (biology) ,Mesenchymal stem cell ,Bioinformatics ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,medicine ,Stem cell ,medicine.symptom ,business ,Confusion - Abstract
Confusion about mesenchymal stem cells is making it easier for people to sell unproven treatments, warn Douglas Sipp, Pamela G. Robey and Leigh Turner. Confusion about mesenchymal stem cells is making it easier for people to sell unproven treatments, warn Douglas Sipp, Pamela G. Robey and Leigh Turner.
- Published
- 2018
- Full Text
- View/download PDF
33. Micro-pellet culture reveals that bone marrow mesenchymal stromal cell (BMSC) chondrogenic induction is triggered by a single day of TGF-β1 exposure
- Author
-
Michael R. Doran, Travis J. Klein, Pamela Gehron Robey, Ross Crawford, and Kathryn Futrega
- Subjects
0303 health sciences ,Stromal cell ,Chemistry ,Growth factor ,medicine.medical_treatment ,Cartilage ,030302 biochemistry & molecular biology ,Mesenchymal stem cell ,Cell fate determination ,Matrix (biology) ,Chondrogenesis ,Cell biology ,03 medical and health sciences ,medicine.anatomical_structure ,medicine ,Bone marrow ,030304 developmental biology - Abstract
Despite immense promise, engineering of stable cartilage tissue from bone marrow-derived stromal cells (BMSCs, also known as bone marrow-derived “mesenchymal stem cells”) remains elusive. Relative cartilage-like matrix deposition is commonly used to guide BMSC chondrogenic optimisation efforts. However, matrix deposition is heterogeneous in most models, and notably, it lags behind cell fate decisions. We reason that the lag time between cell fate decision and matrix accumulation, coupled with matrix heterogeneity, has obscured basic BMSC biological characteristics, such as differentiation kinetics. Here, we utilize a customized microwell platform to assemble hundreds of small-diameter BMSCmicro-pellets and characterized chondrogenic differentiation kinetics in response to the canonical signaling molecule, transforming growth factor-β1 (TGF-β1).Micro-pellets provide a homogeneous readout, and our experimental design accounts for the significant time delay between growth factor signal and deposition of cartilage-like matrix. While 14-to-21-day induction protocols are routine, BMSCmicro-pellet cultures reveal that a single day of TGF-β1 exposure was sufficient to trigger chondrogenic differentiation cascades resulting in outcomes similar tomicro-pellets exposed to TGF-β1 for 21 days. RNA-sequencing analysis demonstrated that one day of TGF-β1 exposure was also sufficient to induce hypertrophic cascades in BMSC, not observed in articular chondrocytes. Refocusing chondrogenic induction optimisation efforts from weeks to the first hours or days of culture, using homogeneous model systems, may benefit efforts to build stable cartilage formed by BMSCs.SignificanceThemacro-pellet model, and assumptions generated using it, have permeated BMSC-based cartilage tissue engineering strategies since the 1990s. Using amicro-pellet model, we show that BMSC chondrogenic kinetics are significantly more rapid than historicalmacro-pellets data suggests, and that BMSC chondrogenic and hypertrophic commitment is instructed by a single day of TGF-β1 exposure. This highly relevant study demonstrates that: (1)macro-pellets, which are large heterogeneous tissue models confound the differentiation kinetics visible inmicro-pellet models; (2) induction strategies should focus on the first hours or days of culture; (3) even a single day of TGF-β1 exposure drives BMSC to form hypertrophic tissuein vivo, requiring early intervention to prevent hypertrophy; and (4) articular chondrocytes and BMSCs respond distinctly to TGF-β1.
- Published
- 2019
- Full Text
- View/download PDF
34. Lineage-specific differentiation of osteogenic progenitors from pluripotent stem cells reveals the FGF1-RUNX2 association in neural crest-derived osteoprogenitors
- Author
-
Byron W H Mui, Vamsee D. Myneni, Pamela Gehron Robey, Madison Hockaday, Randall K. Merling, Katarzyna Futrega, Luis Fernandez de Castro Diaz, Kulsum Iqbal, Fahad Kidwai, Dan S. Kaufman, Janice Lee, Deepika Arora, Sania Ali, Daniel Martin, Natasha Cherman, and Moaz Ahmad
- Subjects
0301 basic medicine ,Male ,Pluripotent Stem Cells ,MAP Kinase Signaling System ,Cellular differentiation ,Organogenesis ,Core Binding Factor Alpha 1 Subunit ,Biology ,Fibroblast growth factor ,Embryonic Stem Cells/Induced Pluripotent Stem Cells ,osteogenesis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Paraxial mesoderm ,Animals ,Humans ,Cell Lineage ,Induced pluripotent stem cell ,Principal Component Analysis ,Lateral plate mesoderm ,Neural crest ,Cell Differentiation ,Cell Biology ,Embryonic stem cell ,Cell biology ,030104 developmental biology ,Neural Crest ,bone development ,embryonic structures ,Molecular Medicine ,Fibroblast Growth Factor 1 ,Transcriptome ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Human pluripotent stem cells (hPSCs) can provide a platform to model bone organogenesis and disease. To reflect the developmental process of the human skeleton, hPSC differentiation methods should include osteogenic progenitors (OPs) arising from three distinct embryonic lineages: the paraxial mesoderm, lateral plate mesoderm, and neural crest. Although OP differentiation protocols have been developed, the lineage from which they are derived, as well as characterization of their genetic and molecular differences, has not been well reported. Therefore, to generate lineage‐specific OPs from human embryonic stem cells and human induced pluripotent stem cells, we employed stepwise differentiation of paraxial mesoderm‐like cells, lateral plate mesoderm‐like cells, and neural crest‐like cells toward their respective OP subpopulation. Successful differentiation, confirmed through gene expression and in vivo assays, permitted the identification of transcriptomic signatures of all three cell populations. We also report, for the first time, high FGF1 levels in neural crest‐derived OPs—a notable finding given the critical role of fibroblast growth factors (FGFs) in osteogenesis and mineral homeostasis. Our results indicate that FGF1 influences RUNX2 levels, with concomitant changes in ERK1/2 signaling. Overall, our study further validates hPSCs' power to model bone development and disease and reveals new, potentially important pathways influencing these processes., Human pluripotent stem cells were stepwise differentiated toward paraxial mesoderm‐, lateral plate mesoderm‐, and neural crest‐derived osteogenic progenitors (OPs). Transcriptomic signatures of these three cell subpopulations were identified, with a new finding of high FGF1 levels and its association with RUNX2 in neural crest‐derived OPs.
- Published
- 2019
35. Erythropoietin modulates bone marrow stromal cell differentiation
- Author
-
Soumyadeep Dey, Luis F de Castro, Pamela Gehron Robey, Constance Tom Noguchi, and Sukanya Suresh
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Histology ,Stromal cell ,Physiology ,Endocrinology, Diabetes and Metabolism ,030209 endocrinology & metabolism ,Bone morphogenetic protein ,Article ,lcsh:Physiology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,hemic and lymphatic diseases ,Internal medicine ,Adipocyte ,medicine ,Bone ,lcsh:QH301-705.5 ,lcsh:QP1-981 ,Chemistry ,Erythropoietin receptor ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,lcsh:Biology (General) ,Erythropoietin ,Adipogenesis ,Cortical bone ,Bone marrow ,Fat metabolism ,medicine.drug - Abstract
Erythropoietin is essential for bone marrow erythropoiesis and erythropoietin receptor on non-erythroid cells including bone marrow stromal cells suggests systemic effects of erythropoietin. Tg6 mice with chronic erythropoietin overexpression have a high hematocrit, reduced trabecular and cortical bone and bone marrow adipocytes, and decreased bone morphogenic protein 2 driven ectopic bone and adipocyte formation. Erythropoietin treatment (1 200 IU·kg–1) for 10 days similarly exhibit increased hematocrit, reduced bone and bone marrow adipocytes without increased osteoclasts, and reduced bone morphogenic protein signaling in the bone marrow. Interestingly, endogenous erythropoietin is required for normal differentiation of bone marrow stromal cells to osteoblasts and bone marrow adipocytes. ΔEpoRE mice with erythroid restricted erythropoietin receptor exhibit reduced trabecular bone, increased bone marrow adipocytes, and decreased bone morphogenic protein 2 ectopic bone formation. Erythropoietin treated ΔEpoRE mice achieved hematocrit similar to wild-type mice without reduced bone, suggesting that bone reduction with erythropoietin treatment is associated with non-erythropoietic erythropoietin response. Bone marrow stromal cells from wild-type, Tg6, and ΔEpoRE-mice were transplanted into immunodeficient mice to assess development into a bone/marrow organ. Like endogenous bone formation, Tg6 bone marrow cells exhibited reduced differentiation to bone and adipocytes indicating that high erythropoietin inhibits osteogenesis and adipogenesis, while ΔEpoRE bone marrow cells formed ectopic bones with reduced trabecular regions and increased adipocytes, indicating that loss of erythropoietin signaling favors adipogenesis at the expense of osteogenesis. In summary, endogenous erythropoietin signaling regulates bone marrow stromal cell fate and aberrant erythropoietin levels result in their impaired differentiation.
- Published
- 2019
- Full Text
- View/download PDF
36. RANKL Inhibition in Fibrous Dysplasia of Bone: A Preclinical Study in a Mouse Model of the Human Disease
- Author
-
Samantha Donsante, Franco Marinozzi, Emanuela Spica, Alessandro Corsi, Alan Boyde, Rossella Labella, Fabiano Bini, Annamaria Di Filippo, Mara Riminucci, Biagio Palmisano, Domenico Raimondo, Cristina Remoli, and Pamela Gehron Robey
- Subjects
musculoskeletal diseases ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Bone pathology ,030209 endocrinology & metabolism ,Mice, Transgenic ,Osteolysis ,Bone resorption ,Bone and Bones ,bone remodelling ,denosumab ,fibrous dysplasia ,RANKL ,GSα ,Bone remodeling ,03 medical and health sciences ,0302 clinical medicine ,Human disease ,Calcification, Physiologic ,Peptide Elongation Factor 1 ,GTP-Binding Protein alpha Subunits, Gs ,Medicine ,Animals ,Humans ,Orthopedics and Sports Medicine ,Bone pain ,biology ,business.industry ,Fibrous dysplasia ,RANK Ligand ,Fibrous Dysplasia of Bone ,medicine.disease ,Biomechanical Phenomena ,Rats ,Disease Models, Animal ,030104 developmental biology ,Denosumab ,Phenotype ,biology.protein ,Disease Progression ,Histopathology ,medicine.symptom ,business ,medicine.drug - Abstract
Fibrous dysplasia of bone/McCune-Albright syndrome (Polyostotic FD/MAS; OMIM#174800) is a crippling skeletal disease caused by gain-of-function mutations of Gs α. Enhanced bone resorption is a recurrent histological feature of FD and a major cause of fragility of affected bones. Previous work suggests that increased bone resorption in FD is driven by RANKL and some studies have shown that the anti-RANKL monoclonal antibody, denosumab, reduces bone turnover and bone pain in FD patients. However, the effect of RANKL inhibition on the histopathology of FD and its impact on the natural history of the disease remain to be assessed. In this study, we treated the EF1α-Gs αR201C mice, which develop an FD-like phenotype, with an anti-mouse RANKL monoclonal antibody. We found that the treatment induced marked radiographic and microscopic changes at affected skeletal sites in 2-month-old mice. The involved skeletal segments became sclerotic due to the deposition of new, highly mineralized bone within developing FD lesions and showed a higher mechanical resistance compared to affected segments from untreated transgenic mice. Similar changes were also detected in older mice with a full-blown skeletal phenotype. The administration of anti-mouse RANKL antibody arrested the growth of established lesions and, in young mice, prevented the appearance of new ones. However, after drug withdrawal, the newly formed bone was remodelled into FD tissue and the disease progression resumed in young mice. Taken together, our results show that the anti-RANKL antibody significantly affected the bone pathology and natural history of FD in the mouse. Pending further work on the prevention and management of relapse after treatment discontinuation, our preclinical study suggests that RANKL inhibition may be an effective therapeutic option for FD patients. © 2019 American Society for Bone and Mineral Research.
- Published
- 2019
37. Intramyocardial Bone Marrow Stem Cells in Patients Undergoing Cardiac Surgical Revascularization
- Author
-
Justin G. Miller, David F. Stroncek, Vandana Sachdev, Pamela Gehron Robey, Andrew E. Arai, Joshua L. Chan, Keith A. Horvath, and Yifu Zhou
- Subjects
Pulmonary and Respiratory Medicine ,Cardiac function curve ,Male ,medicine.medical_specialty ,medicine.medical_treatment ,Myocardial Ischemia ,030204 cardiovascular system & hematology ,Coronary Angiography ,Mesenchymal Stem Cell Transplantation ,Article ,Ventricular Function, Left ,Injections ,Angina ,03 medical and health sciences ,0302 clinical medicine ,stomatognathic system ,Internal medicine ,Preoperative Care ,medicine ,Myocardial Revascularization ,Humans ,Ischemic cardiomyopathy ,Ejection fraction ,business.industry ,Myocardium ,Mesenchymal Stem Cells ,Stem-cell therapy ,Middle Aged ,medicine.disease ,Transmyocardial revascularization ,Myocardial Contraction ,medicine.anatomical_structure ,030228 respiratory system ,Cardiology ,Quality of Life ,Feasibility Studies ,Surgery ,Female ,Bone marrow ,Cardiology and Cardiovascular Medicine ,business ,Artery ,Follow-Up Studies - Abstract
Background Bone marrow stromal or stem cells (BMSCs) remain a promising potential therapy for ischemic cardiomyopathy. The primary objective of this study was to evaluate the safety and feasibility of direct intramyocardial injection of autologous BMSCs in patients undergoing transmyocardial revascularization (TMR) or coronary artery bypass graft surgery (CABG). Methods A phase I trial was conducted on adult patients who had ischemic heart disease with depressed left ventricular ejection fraction and who were scheduled to undergo TMR or CABG. Autologous BMSCs were expanded for 3 weeks before the scheduled surgery. After completion of surgical revascularization, BMSCs were directly injected into ischemic myocardium. Safety and feasibility of therapy were assessed. Cardiac functional status and changes in quality of life were evaluated at 1 year. Results A total of 14 patients underwent simultaneous BMSC and surgical revascularization therapy (TMR+BMSCs = 10; CABG+BMSCs = 4). BMSCs were successfully expanded, and no significant complications occurred as a result of the procedure. Regional contractility in the cell-treated areas demonstrated improvement at 12 months compared with baseline (TMR+BMSCs Δ strain: −4.6% ± 2.1%; P = .02; CABG+MSCs Δ strain: −4.2% ± 6.0%; P = .30). Quality of life was enhanced, with substantial reduction in angina scores at 1 year after treatment (TMR+BMSCs: 1.3 ± 1.2; CABG+MSCs: 1.0 ± 1.4). Conclusions In this phase I trial, direct intramyocardial injection of autologous BMSCs in conjunction with TMR or CABG was technically feasible and could be performed safely. Preliminary results demonstrate improved cardiac function and quality of life in patients at 1 year after treatment.
- Published
- 2019
38. Neonatal McCune‐Albright Syndrome: A Unique Syndromic Profile With an Unfavorable Outcome
- Author
-
Pamela Gehron Robey, Michael T. Collins, Mara Riminucci, David L Donaldson, A. Corsi, and Natasha Cherman
- Subjects
musculoskeletal diseases ,Pathology ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,CHOLESTASIS ,030209 endocrinology & metabolism ,Diseases of the musculoskeletal system ,MCCUNE‐ALBRIGHT SYNDROME ,Postzygotic mutation ,McCune–Albright syndrome ,GNAS ,03 medical and health sciences ,Cushing syndrome ,0302 clinical medicine ,McCune-Albright syndrome ,gnas ,cushing syndrome ,cholestasis ,neonatal ,medicine ,GNAS complex locus ,Precocious puberty ,Orthopedics and Sports Medicine ,Polyostotic fibrous dysplasia ,030304 developmental biology ,Orthopedic surgery ,0303 health sciences ,biology ,Special Issue ,business.industry ,medicine.disease ,NEONATAL ,3. Good health ,Osteopenia ,CUSHING SYNDROME ,RC925-935 ,biology.protein ,Age of onset ,business ,RD701-811 - Abstract
Somatic gain‐of‐function mutations of GNAS cause a spectrum of clinical phenotypes, ranging from McCune‐Albright syndrome (MAS) to isolated disease of bone, endocrine glands, and more rarely, other organs. In MAS, a syndrome classically characterized by polyostotic fibrous dysplasia (FD), café‐au‐lait (CAL) skin spots, and precocious puberty, the heterogenity of organ involvement, age of onset, and clinical severity of the disease are thought to reflect the variable size and the random distribution of the mutated cell clone arising from the postzygotic mutation. We report a case of neonatal MAS with hypercortisolism and cholestatic hepatobiliary dysfunction in which bone changes indirectly emanating from the disease genotype, and distinct from FD, led to a fatal outcome. Pulmonary embolism of marrow and bone fragments secondary to rib fractures was the immediate cause of death. Ribs, and all other skeletal segments, were free of changes of typical FD and fractures appeared to be the result of a mild‐to‐moderate degree of osteopenia. The mutated allele was abundant in the adrenal glands and liver, but not in skin, muscle, and fractured ribs, where it could only be demonstrated using a much more sensitive PNA hybridization probe‐based FRET (Förster resonance energy transfer) technique. Histologically, bilateral adrenal hyperplasia and cholestatic disease matched the abundant disease genotype in the adrenals and liver. Based on this case and other sporadic reports, it appears that gain‐of‐function mutations of GNAS underlie a unique syndromic profile in neonates characterized by CAL skin spots, hypercortisolism, hyperthyroidism, hepatic and cardiac dysfunction, and an absence (or latency) of FD, often with a lethal outcome. Taken together, our and previous cases highlight the phenotypic severity and the diagnostic and therapeutic challenges of MAS in neonates. Furthermore, our case specifically points out how secondary bone changes, unrelated to the direct impact of the mutation, may contribute to the unfavorable outcome of very early‐onset MAS. © 2018 The Authors JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
- Published
- 2019
- Full Text
- View/download PDF
39. 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
- Author
-
Enrico Tagliafico, Cristina Remoli, Pamela Gehron Robey, Elena Tenedini, Stefanie Liedtke, Paolo Bianco, Giulio Cossu, Benedetto Sacchetti, Giuseppe Giannicola, Marta Serafini, Isabella Saggio, Maurilio Sampaolesi, Alessia Funari, Mara Riminucci, and Gesine Kögler
- Subjects
0301 basic medicine ,bone marrow stromal cell ,Cellular differentiation ,Gene Expression ,Biochemistry ,skeletal progenitors ,Transcriptome ,Mice ,Osteogenesis ,lcsh:QH301-705.5 ,mesenchymal stem cell ,lcsh:R5-920 ,Cell Differentiation ,differentiation ,Fetal Blood ,Cell biology ,medicine.anatomical_structure ,Phenotype ,lcsh:Medicine (General) ,Chondrogenesis ,Mesoderm ,Stromal cell ,Satellite Cells, Skeletal Muscle ,in vivo assays ,Transplantation, Heterologous ,Bone Marrow Cells ,Biology ,Mesenchymal Stem Cell Transplantation ,Article ,myogenic progenitors ,03 medical and health sciences ,Genetics ,medicine ,Animals ,Humans ,Cell Lineage ,Progenitor cell ,hematopoietic microenvironment ,Gene Expression Profiling ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Cell Biology ,Transplantation ,030104 developmental biology ,lcsh:Biology (General) ,transplantation ,Developmental Biology ,Immunology ,Microvessels ,Pericytes ,Developmental biology ,Biomarkers - Abstract
Summary A widely shared view reads that mesenchymal stem/stromal cells (“MSCs”) are ubiquitous in human connective tissues, can be defined by a common in vitro phenotype, share a skeletogenic potential as assessed by in vitro differentiation assays, and coincide with ubiquitous pericytes. Using stringent in vivo differentiation assays and transcriptome analysis, we show that human cell populations from different anatomical sources, regarded as “MSCs” based on these criteria and assumptions, actually differ widely in their transcriptomic signature and in vivo differentiation potential. In contrast, they share the capacity to guide the assembly of functional microvessels in vivo, regardless of their anatomical source, or in situ identity as perivascular or circulating cells. This analysis reveals that muscle pericytes, which are not spontaneously osteochondrogenic as previously claimed, may indeed coincide with an ectopic perivascular subset of committed myogenic cells similar to satellite cells. Cord blood-derived stromal cells, on the other hand, display the unique capacity to form cartilage in vivo spontaneously, in addition to an assayable osteogenic capacity. These data suggest the need to revise current misconceptions on the origin and function of so-called “MSCs,” with important applicative implications. The data also support the view that rather than a uniform class of “MSCs,” different mesoderm derivatives include distinct classes of tissue-specific committed progenitors, possibly of different developmental origin., Highlights • CD146+ “MSCs” from different tissues exhibit different transcriptional profiles • CD146+ “MSCs” from different tissues have different differentiation capacities • CD146+ “MSCs” from different tissues organize blood vessels and become pericytes, Bianco, Riminucci, Robey, and colleagues have provided evidence that “mesenchymal stem/stromal cells” (according to current “jargon”), derived from different sources (bone marrow, muscle, cord blood) vary widely in their transcriptional profile, and their differentiation capacity as assessed by in vitro assays and in vivo transplantation assays. Bone marrow “MSCs” form bone and support hematopoiesis, but are not myogenic; muscle “MSCs” are spontaneously myogenic, but do not form bone; cord blood “MSCs” are inherently chondrogenic, and do form bone, but do not support hematopoiesis. However, despite their significant differences, “MSCs” from different sources are capable of forming pericytes when co-transplanted with endothelial cells in vivo, resulting in the development of functional blood vessels. These findings are important not only in understanding the biology of specific tissues, but also from an applicative clinical angle.
- Published
- 2016
- Full Text
- View/download PDF
40. Bi-allelic CSF1R Mutations Cause Skeletal Dysplasia of Dysosteosclerosis-Pyle Disease Spectrum and Degenerative Encephalopathy with Brain Malformation
- Author
-
James Y. Garbern, José Francisco da Silva Franco, Gen Nishimura, Melanie A. Knight, Débora Romeo Bertola, Asako Takanohashi, Raphael Schiffmann, Chong Ae Kim, Maria Rita Passos-Bueno, Rachel Sayuri Honjo, Kinya Ishikawa, Pelin Ozlem Simsek-Kiper, Margaret Timmons, Yuko Segawa, Hirofumi Ohashi, Kenneth H. Fischbeck, Cas Simons, Takanori Yokota, Long Guo, Alan Boyde, Carlos Ferreira, Noriko Miyake, Shiro Ikegawa, Zheng Wang, J. Spranger, Guilherme L. Yamamoto, Adeline Vanderver, Asuka Saito, Yoichiro Nishida, Naomichi Matsumoto, Andrew B. Singleton, Camila Manso Musso, Ryan J. Taft, Bryan R. Lajoie, Amy Pizzino, Pamela Gehron Robey, Li Yan, and Satoru Ishibashi
- Subjects
0301 basic medicine ,Adult ,Male ,Pathology ,medicine.medical_specialty ,Lineage (genetic) ,Adolescent ,Biology ,Compound heterozygosity ,medicine.disease_cause ,Osteochondrodysplasias ,Leukoencephalopathy ,Colony stimulating factor 1 receptor ,03 medical and health sciences ,Mice ,Young Adult ,0302 clinical medicine ,Leukoencephalopathies ,Report ,Genetics ,medicine ,Animals ,Humans ,Genetics (clinical) ,Alleles ,Mice, Knockout ,Mutation ,Brain ,medicine.disease ,Metaphyseal dysplasia ,030104 developmental biology ,Phenotype ,Dysplasia ,Receptors, Granulocyte-Macrophage Colony-Stimulating Factor ,Child, Preschool ,Hereditary diffuse leukoencephalopathy with spheroids ,Female ,030217 neurology & neurosurgery ,Osteosclerosis - Abstract
Colony stimulating factor 1 receptor (CSF1R) plays key roles in regulating development and function of the monocyte/macrophage lineage, including microglia and osteoclasts. Mono-allelic mutations of CSF1R are known to cause hereditary diffuse leukoencephalopathy with spheroids (HDLS), an adult-onset progressive neurodegenerative disorder. Here, we report seven affected individuals from three unrelated families who had bi-allelic CSF1R mutations. In addition to early-onset HDLS-like neurological disorders, they had brain malformations and skeletal dysplasia compatible to dysosteosclerosis (DOS) or Pyle disease. We identified five CSF1R mutations that were homozygous or compound heterozygous in these affected individuals. Two of them were deep intronic mutations resulting in abnormal inclusion of intron sequences in the mRNA. Compared with Csf1r-null mice, the skeletal and neural phenotypes of the affected individuals appeared milder and variable, suggesting that at least one of the mutations in each affected individual is hypomorphic. Our results characterized a unique human skeletal phenotype caused by CSF1R deficiency and implied that bi-allelic CSF1R mutations cause a spectrum of neurological and skeletal disorders, probably depending on the residual CSF1R function.
- Published
- 2018
41. Pluripotent Stem Cell Platforms for Drug Discovery
- Author
-
Michael M. Gottesman, Wei Zheng, Kevin G. Chen, Kye-Yoon Park, Barbara S. Mallon, Ronald D.G. McKay, and Pamela Gehron Robey
- Subjects
0301 basic medicine ,Drug ,Pluripotent Stem Cells ,Cystic Fibrosis ,media_common.quotation_subject ,Cell Culture Techniques ,Cystic Fibrosis Transmembrane Conductance Regulator ,ATP-binding cassette transporter ,Computational biology ,Biology ,Article ,03 medical and health sciences ,Drug Development ,Drug Discovery ,Animals ,Humans ,Molecular Targeted Therapy ,Induced pluripotent stem cell ,Molecular Biology ,media_common ,Drug discovery ,Cystic fibrosis transmembrane conductance regulator ,Organoids ,Drug repositioning ,030104 developmental biology ,Drug development ,Cell culture ,biology.protein ,Molecular Medicine ,ATP-Binding Cassette Transporters - Abstract
Use of human pluripotent stem cells (hPSCs) and their differentiated derivatives have led to recent proof-of-principle drug discoveries, defining a pathway to the implementation of hPSC-based drug discovery (hPDD). Current hPDD strategies, however, have inevitable conceptual biases and technological limitations, including the dimensionality of cell-culture methods, cell maturity and functionality, experimental variability, and data reproducibility. In this review, we dissect representative hPDD systems via analysis of hPSC-based 2D-monolayers, 3D culture, and organoids. We discuss mechanisms of drug discovery and drug repurposing, and roles of membrane drug transporters in tissue maturation and hPDD using the example of drugs that target various mutations of CFTR, the cystic fibrosis transmembrane conductance regulator gene, in patients with cystic fibrosis.
- Published
- 2018
42. Human bone cellsin vitro
- Author
-
John D. Termine and Pamela Gehron Robey
- Subjects
biology ,Endocrinology, Diabetes and Metabolism ,Osteoblast ,Ascorbic acid ,Extracellular matrix ,Endocrinology ,medicine.anatomical_structure ,Biochemistry ,Proteoglycan ,Cell culture ,medicine ,biology.protein ,Alkaline phosphatase ,Orthopedics and Sports Medicine ,Osteonectin ,Type I collagen - Abstract
Human bone cell cultures were established by maintaining collagenase-treated, bone fragments in low Ca++ medium. The resulting cell cultures exhibited a high level of alkaline phosphatase activity and produced a significant increase in intracellular cAMP when exposed to the 1-34 fragment of human parathyroid hormone. With continued culture, the cells formed a thick, extracellular matrix that mineralized when cultures were provided daily with normal levels of calcium, fresh ascorbic acid (50 μg/ml) and 10 mM β-glycerol phosphate. Biosynthetically, these cells produced type I collagen (without any type III collagen), and the bone-specific protein, osteonectin. In addition, the cells produced sulfated macromolecules electrophoretically identical to those positively identified as the bone proteoglycan in parallel cultures of fetal bovine bone cells. This technique provides a useful system for the study of osteoblast metabolismin vitro.
- Published
- 2018
43. Skeletal Stem Cells/Bone Marrow Stromal Cells
- Author
-
Pamela Gehron Robey
- Subjects
Stromal cell ,medicine.anatomical_structure ,Hematopoietic stem cell niche ,Immunology ,medicine ,Clinical uses of mesenchymal stem cells ,Bone marrow ,Stem cell ,Biology ,Bone regeneration ,Adult stem cell ,Stem cell transplantation for articular cartilage repair ,Cell biology - Abstract
It is now clear that the bone/marrow organ contains a multipotent skeletal stem cell (SSC) that is able to recreate cartilage, bone, hematopoiesis-supportive stroma, and marrow adipocytes. Based on their ability to form bone, and to control osteoclastogenesis (at least in part), they are central mediators of skeletal homeostasis. As such, mutations that affect their biological activities can have a profound effect on the skeleton, and even on hematopoiesis due to their participation in the hematopoietic stem cell niche. Furthermore, the fate choices of SSCs in the postnatal organism into osteogenic and adipogenic progeny are highly influenced by changes in the microenvironment in which they reside, and are mediated by numerous signaling pathways, genomic, and epigenetic processes. Lastly, SSCs are an essential ingredient for any process aimed at enduring regeneration, bone regeneration by the cells themselves due to their ability to mediate bone turnover.
- Published
- 2018
- Full Text
- View/download PDF
44. Impaired function of bone marrow stromal cells in systemic mastocytosis
- Author
-
Miklos Krepuska, Todd M. Wilson, David M. Stroncek, Eva Mezey, Yun Bai, Pamela Gehron Robey, Dean D. Metcalfe, Jiaqiang Ren, Krisztián Németh, and Marianna Sabatino
- Subjects
Adult ,Male ,Stromal cell ,CD34 ,Bone Marrow Cells ,Biology ,Article ,Bone remodeling ,Colony-Forming Units Assay ,Mice ,Mastocytosis, Systemic ,stomatognathic system ,Osteogenesis ,medicine ,Animals ,Humans ,Gene Regulatory Networks ,Systemic mastocytosis ,Cell Shape ,lcsh:QH301-705.5 ,Aged ,Cell Proliferation ,Medicine(all) ,Adipogenesis ,Gene Expression Profiling ,Mesenchymal stem cell ,hemic and immune systems ,General Medicine ,Cell Biology ,Middle Aged ,medicine.disease ,Tissue Donors ,Hematopoiesis ,Proto-Oncogene Proteins c-kit ,Haematopoiesis ,medicine.anatomical_structure ,lcsh:Biology (General) ,Case-Control Studies ,Mutation ,Immunology ,Cancer research ,Female ,Bone marrow ,Stromal Cells ,Stem cell ,Developmental Biology - Abstract
Patients with systemic mastocytosis (SM) have a wide variety of problems, including skeletal abnormalities. The disease results from a mutation of the stem cell receptor (c-kit) in mast cells and we wondered if the function of bone marrow stromal cells (BMSCs; also known as MSCs or mesenchymal stem cells) might be affected by the invasion of bone marrow by mutant mast cells. As expected, BMSCs from SM patients do not have a mutation in c-kit, but they proliferate poorly. In addition, while osteogenic differentiation of the BMSCs seems to be deficient, their adipogenic potential appears to be increased. Since the hematopoietic supportive abilities of BMSCs are also important, we also studied the engraftment in NSG mice of human CD34+ hematopoietic progenitors, after being co-cultured with BMSCs of healthy volunteers vs. BMSCs derived from patients with SM. BMSCs derived from the bone marrow of patients with SM could not support hematopoiesis to the extent that healthy BMSCs do. Finally, we performed an expression analysis and found significant differences between healthy and SM derived BMSCs in the expression of genes with a variety of functions, including the WNT signaling, ossification, and bone remodeling. We suggest that some of the symptoms associated with SM might be driven by epigenetic changes in BMSCs caused by dysfunctional mast cells in the bone marrow of the patients.
- Published
- 2015
- Full Text
- View/download PDF
45. Osteoblast-Specific Expression of the Fibrous Dysplasia (FD)-Causing MutationGsαR201CProduces a High Bone Mass Phenotype but Does Not Reproduce FD in the Mouse
- Author
-
Graham R. Davis, Emanuela Spica, Alberto Di Consiglio, Isabella Saggio, Kenn Holmbeck, Stefano Michienzi, Ana Cumano, Alan Boyde, Stefania Cersosimo, Mara Riminucci, Cristina Remoli, Pamela Gehron Robey, Benedetto Sacchetti, and Paolo Bianco
- Subjects
0303 health sciences ,Pathology ,medicine.medical_specialty ,Stromal cell ,Endocrinology, Diabetes and Metabolism ,Fibrous dysplasia ,030209 endocrinology & metabolism ,Osteoblast ,Biology ,medicine.disease ,Phenotype ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Bone cell ,medicine ,Orthopedics and Sports Medicine ,Cortical bone ,Bone marrow ,Stem cell ,030304 developmental biology - Abstract
We recently reported the generation and initial characterization of the first direct model of human fibrous dysplasia (FD; OMIM #174800), obtained through the constitutive systemic expression of one of the disease-causing mutations, Gsα(R201C) , in the mouse. To define the specific pathogenetic role(s) of individual cell types within the stromal/osteogenic system in FD, we generated mice expressing Gsα(R201C) selectively in mature osteoblasts using the 2.3kb Col1a1 promoter. We show here that this results in a striking high bone mass phenotype but not in a mimicry of human FD. The high bone mass phenotype involves specifically a deforming excess of cortical bone and prolonged and ectopic cortical bone remodeling. Expression of genes characteristic of late stages of bone cell differentiation/maturation is profoundly altered as a result of expression of Gsα(R201C) in osteoblasts, and expression of the Wnt inhibitor Sost is reduced. Although high bone mass is, in fact, a feature of some types/stages of FD lesions in humans, it is marrow fibrosis, localized loss of adipocytes and hematopoietic tissue, osteomalacia, and osteolytic changes that together represent the characteristic pathological profile of FD, as well as the sources of specific morbidity. None of these features are reproduced in mice with osteoblast-specific expression of Gsα(R201C) . We further show that hematopoietic progenitor/stem cells, as well as more mature cell compartments, and adipocyte development are normal in these mice. These data demonstrate that effects of Gsα mutations underpinning FD-defining tissue changes and morbidity do not reflect the effects of the mutations on osteoblasts proper. © 2015 American Society for Bone and Mineral Research. © 2014 American Society for Bone and Mineral Research.
- Published
- 2015
- Full Text
- View/download PDF
46. Mice Deficient inAKAP13(BRX) Are Osteoporotic and Have Impaired Osteogenesis
- Author
-
Tiffany Chu, Caroline Quaglieri, Marian F. Young, Hisashi Koide, Julian C. Lui, Ichiro Tatsuno, James H. Segars, Xiaoxiao C Guo, Jeffrey Baron, Tomoshige Kino, Kenn Holmbeck, Paul H. Driggers, and Pamela Gehron Robey
- Subjects
Bone growth ,medicine.medical_specialty ,RHOA ,biology ,Chemistry ,Endocrinology, Diabetes and Metabolism ,Osteoporosis ,Bone Marrow Stem Cell ,Osteoblast ,medicine.disease ,Bone remodeling ,RUNX2 ,Endocrinology ,medicine.anatomical_structure ,Internal medicine ,medicine ,biology.protein ,Alkaline phosphatase ,Orthopedics and Sports Medicine - Abstract
Mechanical stimulation is crucial to bone growth and triggers osteogenic differentiation through a process involving Rho and protein kinase A. We previously cloned a gene (AKAP13, aka BRX) encoding a protein kinase A-anchoring protein in the N-terminus, a guanine nucleotide-exchange factor for RhoA in the mid-section, coupled to a carboxyl region that binds to estrogen and glucocorticoid nuclear receptors. Because of the critical role of Rho, estrogen, and glucocorticoids in bone remodeling, we examined the multifunctional role of Akap13. Akap13 was expressed in bone, and mice haploinsufficient for Akap13 (Akap13+/–) displayed reduced bone mineral density, reduced bone volume/total volume, and trabecular number, and increased trabecular spacing; resembling the changes observed in osteoporotic bone. Consistent with the osteoporotic phenotype, Colony forming unit-fibroblast numbers were diminished in Akap13+/– mice, as were osteoblast numbers and extracellular matrix production when compared to control littermates. Transcripts of Runx2, an essential transcription factor for the osteogenic lineage, and alkaline phosphatase (Alp), an indicator of osteogenic commitment, were both reduced in femora of Akap13+/– mice. Knockdown of Akap13 reduced levels of Runx2 and Alp transcripts in immortalized bone marrow stem cells. These findings suggest that Akap13 haploinsufficient mice have a deficiency in early osteogenesis with a corresponding reduction in osteoblast number, but no impairment of mature osteoblast activity. © 2015 American Society for Bone and Mineral Research.
- Published
- 2015
- Full Text
- View/download PDF
47. p53 Loss Increases the Osteogenic Differentiation of Bone Marrow Stromal Cells
- Author
-
Wendy Dubois, Chand Khanna, Ling Ren, Luis F de Castro, Min Hwa Shin, Jing Huang, Ashish Lal, Maxwell P. Lee, Shunlin Jiang, Glenn Merlino, Yunlong He, Pravin J. Mishra, Hongfeng Gou, Howard H. Yang, and Pamela Gehron Robey
- Subjects
musculoskeletal diseases ,Stromal cell ,Cellular differentiation ,Mesenchymal stem cell ,Cell Biology ,Biology ,Chondrogenesis ,medicine.disease ,RUNX2 ,medicine.anatomical_structure ,stomatognathic system ,Adipogenesis ,medicine ,Cancer research ,Molecular Medicine ,Osteosarcoma ,Bone marrow ,Developmental Biology - Abstract
The tumor suppressor, p53, plays a critical role in suppressing osteosarcoma. Bone marrow stromal cells (BMSCs, also known as bone marrow-derived mesenchymal stem cells) have been suggested to give rise to osteosarcomas. However, the role of p53 in BMSCs has not been extensively explored. Here, we report that p53 regulates the lineage choice of mouse BMSCs (mBMSCs). Compared to mBMSCs with wild-type p53, mBMSCs deficient in p53 have enhanced osteogenic differentiation, but with similar adipogenic and chondrogenic differentiation. The role of p53 in inhibiting osteogenic lineage differentiation is mainly through the action of Runx2, a master transcription factor required for the osteogenic differentiation of mBMSCs. We find that p53 indirectly represses the expression of Runx2 by activating the microRNA-34 family, which suppresses the translation of Runx2. Since osteosarcoma may derive from BMSCs, we examined whether p53 has a role in the osteogenic differentiation of osteosarcoma cells and found that osteosarcoma cells with p53 deletion have higher levels of Runx2 and faster osteogenic differentiation than those with wild-type p53. A systems biology approach reveals that p53-deficient mBMSCs are more closely related to human osteosarcoma while mBMSCs with wild-type p53 are similar to normal human BMSCs. In summary, our results indicate that p53 activity can influence cell fate specification of mBMSCs, and provide molecular and cellular insights into the observation that p53 loss is associated with increased osteosarcoma incidence. Stem Cells 2015;33:1304–1319
- Published
- 2015
- Full Text
- View/download PDF
48. Skeletal stem cells
- Author
-
Paolo Bianco and Pamela Gehron Robey
- Subjects
endocrine system ,Stromal cell ,Clinical uses of mesenchymal stem cells ,Context (language use) ,Biology ,Bone and Bones ,Stroma ,Development at A Glance ,Terminology as Topic ,Bone cell ,Adipocytes ,medicine ,Perichondrium ,Bone organ ,Humans ,Cell Lineage ,Stem Cell Niche ,Molecular Biology ,Stem cell transplantation for articular cartilage repair ,Bone growth ,Multipotent Stem Cells ,Cartilage ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Anatomy ,Cell biology ,Haematopoiesis ,medicine.anatomical_structure ,Multipotent Stem Cell ,Immunology ,Bone marrow ,Stem cell ,Developmental Biology - Abstract
Most bones develop from mesenchymal condensations giving rise to cartilaginous rudiments via a prechondrogenic blastema. The outermost portion of the chondrogenic blastema later evolves into a perichondrium and further into a primitive periosteum at a time when cartilage cores mature to hypertrophy and the primary centers of ossification are established. Chondroprogenitors and osteoprogenitors within the perichondrium and periosteum allow the growth of cartilage and bone. Once bone growth has ceased, multipotent progenitors capable of giving rise to cartilage, bone, and bone marrow stroma remain in the bone marrow, their ultimate destination. Throughout development and growth, human skeletal stem cells generate 15 kg of bone. Ectopic transplantation of marrow fragments results in the formation of a complete bone organ. The distinct osteogenic potential of marrow as a tissue is ascribed to the nonhematopoietic, stromal component that provides the hematopoietic microenvironment in vivo. Stromal cell strains derived in culture from explanted adherent, clonogenic, fibroblast-like cells, generate bone and cartilage when transplanted in diffusion chambers and generate a complete heterotopic bone organ upon in vivo transplantation in open systems. In these systems, bone-forming cells and the stromal components of the bone marrow (reticular cells and adipocytes) are derived from the donor cell population; whereas hematopoietic cells filling the marrow space originate from host hematopoietic stem cells (HSCs).
- Published
- 2015
- Full Text
- View/download PDF
49. Generation of clinical grade human bone marrow stromal cells for use in bone regeneration
- Author
-
David F. Stroncek, Sergei A. Kuznetsov, Harvey G. Klein, Marianna Sabatino, Pamela Gehron Robey, and Jiaqiang Ren
- Subjects
Quality Control ,Pathology ,medicine.medical_specialty ,Bone Regeneration ,Histology ,Stromal cell ,Physiology ,Endocrinology, Diabetes and Metabolism ,Cell Culture Techniques ,Clinical uses of mesenchymal stem cells ,Avascular necrosis ,Mesenchymal Stem Cell Transplantation ,Article ,Bioreactors ,medicine ,Humans ,Bone regeneration ,Cells, Cultured ,Cell Proliferation ,business.industry ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,medicine.disease ,medicine.anatomical_structure ,Cell culture ,Bone marrow ,Stem cell ,business ,Biomarkers - Abstract
In current orthopaedic practice, there is a need to increase the ability to reconstruct large segments of bone lost due to trauma, resection of tumors and skeletal deformities, or when normal regenerative processes have failed such as in non-unions and avascular necrosis. Bone marrow stromal cells (BMSCs, also known as bone marrow-derived mesenchymal stem cells), when used in conjunction with appropriate carriers, represent a means by which to achieve bone regeneration in such cases. While much has been done at the bench and in pre-clinical studies, moving towards clinical application requires the generation of clinical grade cells. What is described herein is an FDA-approved cell manufacturing procedure for the ex vivo expansion of high quality, biologically active human BMSCs. This article is part of a Special Issue entitled Stem Cells and Bone.
- Published
- 2015
- Full Text
- View/download PDF
50. Anti-RANKL treatment in a murine model of fibrous dysplasia
- Author
-
Cristina Remoli, Alessandro Corsi, Pamela Gehron Robey, Mara Riminucci, Rossella Labella, Emanuela Spica, and Biagio Palmisano
- Subjects
biology ,business.industry ,Fibrous dysplasia ,General Medicine ,medicine.disease ,030226 pharmacology & pharmacy ,03 medical and health sciences ,0302 clinical medicine ,RANKL ,Murine model ,medicine ,biology.protein ,Cancer research ,030212 general & internal medicine ,business - Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.