Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Regen Ther ; 24: 536-546, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37860130

ABSTRACT

Vertebrates form their skeletal tissues from three distinct origins (the neural crest, paraxial mesoderm, and lateral plate mesoderm) through two distinct modes of ossification (intramembranous and endochondral ossification). Since the paraxial mesoderm generates both intramembranous and endochondral bones, it is thought to give rise to both osteoprogenitors and osteo-chondroprogenitors. However, it remains unclear what directs the paraxial mesoderm-derived cells toward these different fates in distinct skeletal elements during human skeletal development. To answer this question, we need experimental systems that recapitulate paraxial mesoderm-mediated intramembranous and endochondral ossification processes. In this study, we aimed to develop a human pluripotent stem cell (hPSC)-based system that models the human intramembranous ossification process. We found that spheroid culture of the hPSC-derived paraxial mesoderm derivatives generates osteoprogenitors or osteo-chondroprogenitors depending on stimuli. The former induced intramembranous ossification, and the latter endochondral ossification, in mouse renal capsules. Transcriptional profiling supported the notion that bone signatures were enriched in the intramembranous bone-like tissues. Thus, we developed a system that recapitulates intramembranous ossification, and that enables the induction of two distinct modes of ossification by controlling the cell fate of the hPSC-derived paraxial mesoderm derivatives.

2.
Cell Rep ; 42(4): 112276, 2023 04 25.
Article in English | MEDLINE | ID: mdl-36965484

ABSTRACT

Although the skeleton is essential for locomotion, endocrine functions, and hematopoiesis, the molecular mechanisms of human skeletal development remain to be elucidated. Here, we introduce an integrative method to model human skeletal development by combining in vitro sclerotome induction from human pluripotent stem cells and in vivo endochondral bone formation by implanting the sclerotome beneath the renal capsules of immunodeficient mice. Histological and scRNA-seq analyses reveal that the induced bones recapitulate endochondral ossification and are composed of human skeletal cells and mouse circulatory cells. The skeletal cell types and their trajectories are similar to those of human embryos. Single-cell multiome analysis reveals dynamic changes in chromatin accessibility associated with multiple transcription factors constituting cell-type-specific gene-regulatory networks (GRNs). We further identify ZEB2, which may regulate the GRNs in human osteogenesis. Collectively, these results identify components of GRNs in human skeletal development and provide a valuable model for its investigation.


Subject(s)
Multiomics , Pluripotent Stem Cells , Humans , Mice , Animals , Cell Differentiation , Transcription Factors/metabolism , Gene Expression Regulation , Pluripotent Stem Cells/metabolism
3.
BMC Genomics ; 23(1): 14, 2022 Jan 07.
Article in English | MEDLINE | ID: mdl-34991467

ABSTRACT

BACKGROUND: Interferon regulatory factor-8 (IRF8) and nuclear factor-activated T cells c1 (NFATc1) are two transcription factors that have an important role in osteoclast differentiation. Thanks to ChIP-seq technology, scientists can now estimate potential genome-wide target genes of IRF8 and NFATc1. However, finding target genes that are consistently up-regulated or down-regulated across different studies is hard because it requires analysis of a large number of high-throughput expression studies from a comparable context. METHOD: We have developed a machine learning based method, called, Cohort-based TF target prediction system (cTAP) to overcome this problem. This method assumes that the pathway involving the transcription factors of interest is featured with multiple "functional groups" of marker genes pertaining to the concerned biological process. It uses two notions, Gene-Present Sufficiently (GP) and Gene-Absent Insufficiently (GA), in addition to log2 fold changes of differentially expressed genes for the prediction. Target prediction is made by applying multiple machine-learning models, which learn the patterns of GP and GA from log2 fold changes and four types of Z scores from the normalized cohort's gene expression data. The learned patterns are then associated with the putative transcription factor targets to identify genes that consistently exhibit Up/Down gene regulation patterns within the cohort. We applied this method to 11 publicly available GEO data sets related to osteoclastgenesis. RESULT: Our experiment identified a small number of Up/Down IRF8 and NFATc1 target genes as relevant to osteoclast differentiation. The machine learning models using GP and GA produced NFATc1 and IRF8 target genes different than simply using a log2 fold change alone. Our literature survey revealed that all predicted target genes have known roles in bone remodeling, specifically related to the immune system and osteoclast formation and functions, suggesting confidence and validity in our method. CONCLUSION: cTAP was motivated by recognizing that biologists tend to use Z score values present in data sets for the analysis. However, using cTAP effectively presupposes assembling a sizable cohort of gene expression data sets within a comparable context. As public gene expression data repositories grow, the need to use cohort-based analysis method like cTAP will become increasingly important.


Subject(s)
Osteoclasts , RANK Ligand , Cell Differentiation , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Machine Learning , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Osteoclasts/metabolism , RANK Ligand/metabolism , T-Lymphocytes/metabolism
4.
JBMR Plus ; 6(1): e10570, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35079678

ABSTRACT

Albright hereditary osteodystrophy (AHO) is caused by heterozygous inactivation of GNAS, a complex locus that encodes the alpha-stimulatory subunit of heterotrimeric G proteins (Gsα) in addition to NESP55 and XLαs due to alternative first exons. AHO skeletal manifestations include brachydactyly, brachymetacarpia, compromised adult stature, and subcutaneous ossifications. AHO patients with maternally-inherited GNAS mutations develop pseudohypoparathyroidism type 1A (PHP1A) with resistance to multiple hormones that mediate their actions through G protein-coupled receptors (GPCRs) requiring Gsα (eg, parathyroid hormone [PTH], thyroid-stimulating hormone [TSH], growth hormone-releasing hormone [GHRH], calcitonin) and severe obesity. Paternally-inherited GNAS mutations cause pseudopseudohypoparathyroidism (PPHP), in which patients have AHO skeletal features but do not develop hormonal resistance or marked obesity. These differences between PHP1A and PPHP are caused by tissue-specific reduction of paternal Gsα expression. Previous reports in mice have shown loss of Gsα causes osteopenia due to impaired osteoblast number and function and suggest that AHO patients could display evidence of reduced bone mineral density (BMD). However, we previously demonstrated PHP1A patients display normal-increased BMD measurements without any correlation to body mass index or serum PTH. Due to these observed differences between PHP1A and PPHP, we utilized our laboratory's AHO mouse model to address whether Gsα heterozygous inactivation differentially affects bone remodeling based on the parental inheritance of the mutation. We identified fundamental distinctions in bone remodeling between mice with paternally-inherited (GnasE1+/-p) versus maternally-inherited (GnasE1+/-m) mutations, and these findings were observed predominantly in female mice. Specifically, GnasE1+/-p mice exhibited reduced bone parameters due to impaired bone formation and enhanced bone resorption. GnasE1+/-m mice, however, displayed enhanced bone parameters due to both increased osteoblast activity and normal bone resorption. These in vivo distinctions in bone remodeling between GnasE1+/-p and GnasE1+/-m mice could potentially be related to changes in the bone microenvironment driven by calcitonin-resistance within GnasE1+/-m osteoclasts. Further studies are warranted to assess how Gsα influences osteoblast-osteoclast coupling. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

5.
NPJ Regen Med ; 6(1): 34, 2021 Jun 11.
Article in English | MEDLINE | ID: mdl-34117259

ABSTRACT

The osteogenic potential of bone marrow mesenchymal stem cells (BMSCs) declines dramatically with aging. By using a calvarial defect model, we showed that a senolytic cocktail (dasatinib+quercetin; D + Q) improved osteogenic capacity of aged BMSC both in vitro and in vivo. The study presented a model to assess strategies to improve bone-forming potential on aged BMSCs. D + Q might hold promise for improving BMSC function in aged populations.

6.
Bone ; 144: 115688, 2021 03.
Article in English | MEDLINE | ID: mdl-33065355

ABSTRACT

The IMPC/KOMP program provides the opportunity to screen mice harboring well defined gene-inactivation mutations in a uniform genetic background. The program performs a global tissue phenotyping survey that includes skeletal x-rays and bone density measurements. Because of the relative insensitivity of the two screening tests for detecting variance in bone architecture, we initiated a secondary screen based on µCT and a cryohistolomorphological workflow that was performed on the femur and vertebral compartments on 220 randomly selected knockouts (KOs) and 36 control bone samples over a 2 1/2 year collection period provided by one of the production/phenotyping centers. The performance of the screening protocol was designed to balance throughput and cost versus sensitivity and informativeness such that the output would be of value to the skeletal biology community. Here we report the reliability of this screening protocol to establish criteria for control skeletal variance at the architectural, dynamic and cellular histomorphometric level. Unexpected properties of the control population include unusually high variance in BV/TV in male femurs and greater bone formation and bone turnover rates in the female femur and vertebral trabeculae bone compartments. However, the manner for maintaining bone formation differed between these two bone sites. The vertebral compartment relies on maintaining a greater number of bone forming surfaces while the femoral compartment utilized more matrix production per cell. The comparison of the architectural properties obtained by µCT and histomorphology revealed significant differences in values for BV/TV, Tb.Th and Tb.N which is attributable to sampling density of the two methods. However, as a screening tool, expressing the ratio of KO to the control line as obtained by either method was remarkably similar. It identified KOs with significant variance which led to a more detailed histological analysis. Our findings are exemplified by the Efna4 KO, in which a high BV/TV was identified by µCT and confirmed by histomorphometry in the femur but not in the vertebral compartment. Dynamic labeling showed a marked increase in BFR which was attributable to increased labeling surfaces. Cellular analysis confirmed partitioning of osteoblast to labeling surfaces and a marked decrease in osteoclastic activity on both labeling and quiescent surfaces. This pattern of increased bone modeling would not be expected based on prior studies of the Ephrin-Ephrin receptor signaling pathways between osteoblasts and osteoclasts. Overall, our findings underscore why unbiased screening is needed because it can reveal unknown or unanticipated genes that impact skeletal variation.


Subject(s)
Bone Density , Femur , Animals , Bone and Bones/diagnostic imaging , Computers , Female , Femur/diagnostic imaging , Male , Mice , Reproducibility of Results
7.
Bone ; 142: 115677, 2021 01.
Article in English | MEDLINE | ID: mdl-33022452

ABSTRACT

Chromatin modifying enzymes play essential roles in skeletal development and bone maintenance, and deregulation of epigenetic mechanisms can lead to skeletal growth and malformation disorders. Here, we report a novel skeletal dysplasia phenotype in mice with conditional loss of Disruptor of telomeric silencing 1-like (Dot1L) histone methyltransferase in limb mesenchymal progenitors and downstream descendants. Phenotypic characterizations of mice with Dot1L inactivation by Prrx1-Cre (Dot1L-cKOPrrx1) revealed limb shortening, abnormal bone morphologies, and forelimb dislocations. Our in vivo and in vitro data support a crucial role for Dot1L in regulating growth plate chondrocyte proliferation and differentiation, extracellular matrix production, and secondary ossification center formation. Micro-computed tomography analysis of femurs revealed that partial loss of Dot1L expression is sufficient to impair trabecular bone formation and microarchitecture in young mice. Moreover, RNAseq analysis of Dot1L deficient chondrocytes implicated Dot1L in the regulation of key genes and pathways necessary to promote cell cycle regulation and skeletal growth. Collectively, our data show that early expression of Dot1L in limb mesenchyme provides essential regulatory control of endochondral bone morphology, growth, and stability.


Subject(s)
Chondrocytes , Mesoderm , Animals , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Mice , Phenotype , X-Ray Microtomography
8.
ACS Appl Mater Interfaces ; 12(16): 18235-18249, 2020 Apr 22.
Article in English | MEDLINE | ID: mdl-32212615

ABSTRACT

As one of the major challenges in the field of tissue engineering, large skeletal defects have attracted wide attention from researchers. Collagen (Col) and hydroxyapatite (HA), the most abundant protein and the main component in natural bone, respectively, are usually used as a biomimetic composite material in tissue engineering due to their excellent biocompatibility and biodegradability. In this study, novel intrafibrillar mineralized Col-HA-based scaffolds, constructed in either cellular or lamellar microstructures, were established through a biomimetic method to enhance the new bone-regenerating capability of tissue engineering scaffolds. Moreover, iron (Fe) and manganese (Mn), two of the essential trace elements in the body, were successfully incorporated into the lamellar scaffold to further improve the osteoinductivity of these biomaterials. It was found that the lamellar scaffolds demonstrated better osteogenic abilities compared to both in-house and commercial Col-HA-based cellular scaffolds in vitro and in vivo. Meanwhile, Fe/Mn incorporation further amplified the osteogenic promotion of the lamellar scaffolds. More importantly, a synergistic effect was observed in the Fe and Mn dual-element-incorporated lamellar scaffolds for both in vitro osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and in vivo bone regeneration loaded with fresh bone marrow cells. This study provides a simple but practical strategy for the creation of functional scaffolds for bone regeneration.


Subject(s)
Bone Regeneration/drug effects , Collagen , Durapatite , Tissue Engineering/methods , Tissue Scaffolds/chemistry , 3T3 Cells , Animals , Cell Differentiation/drug effects , Cells, Cultured , Collagen/chemistry , Collagen/pharmacology , Durapatite/chemistry , Durapatite/pharmacology , Mesenchymal Stem Cells/drug effects , Mice , Skull/drug effects , Skull/pathology
9.
Regen Ther ; 14: 19-31, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31988991

ABSTRACT

Clinically relevant human induced pluripotent stem cell (hiPSC) derivatives require efficient protocols to differentiate hiPSCs into specific lineages. Here we developed a fully defined xeno-free strategy to direct hiPSCs toward osteoblasts within 21 days. The strategy successfully achieved the osteogenic induction of four independently derived hiPSC lines by a sequential use of combinations of small-molecule inducers. The induction first generated mesodermal cells, which subsequently recapitulated the developmental expression pattern of major osteoblast genes and proteins. Importantly, Col2.3-Cherry hiPSCs subjected to this strategy strongly expressed the cherry fluorescence that has been observed in bone-forming osteoblasts in vivo. Moreover, the protocol combined with a three-dimensional (3D) scaffold was suitable for the generation of a xeno-free 3D osteogenic system. Thus, our strategy offers a platform with significant advantages for bone biology studies and it will also contribute to clinical applications of hiPSCs to skeletal regenerative medicine.

10.
Aging Cell ; 19(3): e13106, 2020 03.
Article in English | MEDLINE | ID: mdl-31971661

ABSTRACT

Adipose-derived mesenchymal stem cell (ADSC)-based regenerative therapies have shown potential for use in many chronic diseases. Aging diminishes stem cell regenerative potential, yet it is unknown whether stem cells from aged donors cause adverse effects in recipients. ADSCs can be obtained using minimally invasive approaches and possess low immunogenicity. Nevertheless, we found that transplanting ADSCs from old donors, but not those from young donors, induces physical dysfunction in older recipient mice. Using single-cell transcriptomic analysis, we identified a naturally occurring senescent cell-like population in ADSCs primarily from old donors that resembles in vitro-generated senescent cells with regard to a number of key pathways. Our study reveals a previously unrecognized health concern due to ADSCs from old donors and lays the foundation for a new avenue of research to devise interventions to reduce harmful effects of ADSCs from old donors.


Subject(s)
Frailty/etiology , Mesenchymal Stem Cell Transplantation/adverse effects , Mesenchymal Stem Cell Transplantation/methods , Tissue Donors , Transplant Recipients , Age Factors , Animals , Biomarkers , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Longevity/genetics , Male , Mice , Mice, Inbred C57BL , Physical Endurance , Regenerative Medicine/methods , Single-Cell Analysis/methods , Transcriptome , Walking Speed
11.
Ann Biomed Eng ; 48(3): 992-1005, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31037444

ABSTRACT

Despite its regenerative ability, long and segmental bone defect repair remains a significant orthopedic challenge. Conventional tissue engineering efforts induce bone formation through intramembranous ossification (IO) which limits vascular formation and leads to poor bone regeneration. To overcome this challenge, a novel hybrid matrix comprised of a load-bearing polymer template and a gel phase is designed and assessed for bone regeneration. Our previous studies developed a synthetic ECM, hyaluronan (HA)-fibrin (FB), that is able to mimic cartilage-mediated bone formation in vitro. In this study, the well-characterized HA-FB hydrogel is combined with a biodegradable polymer template to form a hybrid matrix. In vitro evaluation of the matrix showed cartilage template formation, cell recruitment and recruited cell osteogenesis, essential stages in endochondral ossification. A transgenic reporter-mouse critical-defect model was used to evaluate the bone healing potential of the hybrid matrix in vivo. The results demonstrated host cell recruitment into the hybrid matrix that led to new bone formation and subsequent remodeling of the mineralization. Overall, the study developed and evaluated a novel load-bearing graft system for bone regeneration via endochondral ossification.


Subject(s)
Bone Regeneration , Mesenchymal Stem Cells/physiology , Osteogenesis , Skull/physiology , Tissue Engineering/methods , Animals , Cells, Cultured , Extracellular Matrix , Fibrin , Humans , Hyaluronic Acid , Hydrogels , Mice, SCID , Porosity
12.
Nat Mater ; 18(6): 627-637, 2019 06.
Article in English | MEDLINE | ID: mdl-31114073

ABSTRACT

Cells are transplanted to regenerate an organs' parenchyma, but how transplanted parenchymal cells induce stromal regeneration is elusive. Despite the common use of a decellularized matrix, little is known as to the pivotal signals that must be restored for tissue or organ regeneration. We report that Alx3, a developmentally important gene, orchestrated adult parenchymal and stromal regeneration by directly transactivating Wnt3a and vascular endothelial growth factor. In contrast to the modest parenchyma formed by native adult progenitors, Alx3-restored cells in decellularized scaffolds not only produced vascularized stroma that involved vascular endothelial growth factor signalling, but also parenchymal dentin via the Wnt/ß-catenin pathway. In an orthotopic large-animal model following parenchyma and stroma ablation, Wnt3a-recruited endogenous cells regenerated neurovascular stroma and differentiated into parenchymal odontoblast-like cells that extended the processes into newly formed dentin with a structure-mechanical equivalency to native dentin. Thus, the Alx3-Wnt3a axis enables postnatal progenitors with a modest innate regenerative capacity to regenerate adult tissues. Depleted signals in the decellularized matrix may be reinstated by a developmentally pivotal gene or corresponding protein.


Subject(s)
Homeodomain Proteins/metabolism , Parenchymal Tissue/physiology , Tooth/cytology , Tooth/embryology , Adolescent , Animals , Female , Homeodomain Proteins/genetics , Humans , Incisor/cytology , Incisor/embryology , Mice, Inbred Strains , Molar, Third/cytology , Organ Culture Techniques , Parenchymal Tissue/cytology , Pregnancy , Promoter Regions, Genetic , Regeneration , Stromal Cells/physiology , Swine , Vascular Endothelial Growth Factor A/genetics , Wnt3A Protein/genetics , Wnt3A Protein/metabolism
13.
J Histochem Cytochem ; 67(6): 401-417, 2019 06.
Article in English | MEDLINE | ID: mdl-30848692

ABSTRACT

The effectiveness of autologous cell-based skeletal repair continues to be controversial in part because in vitro predictors of in vivo human bone formation by cultured human progenitor cells are not reliable. To assist in the development of in vivo assays of human osteoprogenitor potential, a fluorescence-based histology of nondecalcified mineralized tissue is presented that provides multiple criteria to distinguish human and host osteoblasts, osteocytes, and accumulated bone matrix in a mouse calvarial defect model. These include detection of an ubiquitously expressed red fluorescent protein reporter by the implanted human cells, antibodies specific to human bone sialoprotein and a human nuclear antigen, and expression of a bone/fibroblast restricted green fluorescent protein reporter in the host tissue. Using low passage bone marrow-derived stromal cells, robust human bone matrix formation was obtained. However, a striking feature is the lack of mouse bone marrow investment and osteoclasts within the human bone matrix. This deficiency may account for the accumulation of a disorganized human bone matrix that has not undergone extensive remodeling. These features, which would not be appreciated by traditional decalcified paraffin histology, indicate the human bone matrix is not undergoing active remodeling and thus the full differentiation potential of the implanted human cells within currently used mouse models is not being realized.


Subject(s)
Osteogenesis , Animals , Humans , Mice , Optical Imaging , Osteoblasts/cytology , Skull/cytology , Skull/diagnostic imaging , Skull/physiology
14.
Nephrol Dial Transplant ; 34(1): 49-62, 2019 01 01.
Article in English | MEDLINE | ID: mdl-29726981

ABSTRACT

Background: Our laboratory has previously demonstrated that Sirt1endo-/- mice show endothelial dysfunction and exaggerated renal fibrosis, whereas mice with silenced endothelial transforming growth factor beta (TGF-ß) signaling are resistant to fibrogenic signals. Considering the fact that the only difference between these mutant mice is confined to the vascular endothelium, this indicates that secreted substances contribute to these contrasting responses. Methods: We performed an unbiased proteomic analysis of the secretome of renal microvascular endothelial cells (RMVECs) isolated from these two mutants. We cultured renal fibroblasts and RMVECs and used microfluidic devices for coculturing. Results: Dickkopf-3 (DKK3), a putative ligand of the Wnt/ß-catenin pathway, was present exclusively in the fibrogenic secretome. In cultured fibroblasts, DKK3 potently induced myofibroblast activation. In addition, DKK3 antagonized effects of DKK1, a known inhibitor of the Wnt pathway, in conversion of fibroblasts to myofibroblasts. In RMVECs, DKK3 induced endothelial-mesenchymal transition and impaired their angiogenic competence. The inhibition of endothelial outgrowth, enhanced myofibroblast formation and endothelial-mesenchymal transition were confirmed in coculture. In reporter DKK3-eGFP × Col3.6-GFPcyan mice, DKK3 was marginally expressed under basal conditions. Adriamycin-induced nephropathy resulted in upregulation of DKK3 expression in tubular and, to a lesser degree, endothelial compartments. Sulindac sulfide was found to exhibit superior Wnt pathway-suppressive action and decreased DKK3 signals and the extent of renal fibrosis. Conclusions: In conclusion, this unbiased proteomic screen of the profibrogenic endothelial secretome revealed DKK3 acting as an agonist of the Wnt pathway, enhancing formation of myofibroblasts and endothelial-mesenchymal transition and impairing angiogenesis. A potent inhibitor of the Wnt pathway, sulindac sulfide, suppressed nephropathy-induced DKK3 expression and renal fibrosis.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Endothelium, Vascular/pathology , Epithelial-Mesenchymal Transition , Fibrosis/pathology , Kidney Diseases/pathology , Proteome/analysis , Receptor, Transforming Growth Factor-beta Type II/physiology , Sirtuin 1/physiology , Animals , Endothelium, Vascular/metabolism , Fibrosis/metabolism , Kidney Diseases/metabolism , Mice , Mice, Knockout , Proteomics , Wnt Signaling Pathway , beta Catenin/metabolism
15.
Tissue Eng Part A ; 25(5-6): 468-486, 2019 03.
Article in English | MEDLINE | ID: mdl-30136616

ABSTRACT

IMPACT STATEMENT: This study represents significant advancement in the use of biomimetic scaffolds to direct zonal osteochondral tissue formation. We describe the use of a novel fluorescent reporter system that enables the real-time evaluation of cellular differentiation in a nondestructive manner. In this study, we use this tool to confirm the osteogenic and chondrogenic capabilities of our scaffold alongside control scaffolds, and use cryohistological methods to probe zone-specific differences in cell and tissue quality. We believe this approach can be widely adopted by others for a variety of biomaterial and cell systems in the development of tissue engineered therapeutics.


Subject(s)
Biocompatible Materials/pharmacology , Cell Differentiation/drug effects , Chondrogenesis/drug effects , Animals , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/metabolism , Collagen Type I/metabolism , Collagen Type II/metabolism , Fluorescence , Gene Expression Regulation/drug effects , Implants, Experimental , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice, Transgenic , Osteogenesis/drug effects , Osteogenesis/genetics , Subcutaneous Tissue/drug effects , Subcutaneous Tissue/physiology , Tissue Scaffolds/chemistry
16.
J Bone Miner Res ; 33(10): 1721-1728, 2018 10.
Article in English | MEDLINE | ID: mdl-30133922

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.


Subject(s)
Biomedical Research , Bone and Bones/physiology , Cell- and Tissue-Based Therapy , Regeneration/physiology , Animals , Disease Models, Animal , Humans , Regenerative Medicine
17.
Curr Osteoporos Rep ; 16(2): 77-94, 2018 04.
Article in English | MEDLINE | ID: mdl-29508144

ABSTRACT

PURPOSE OF REVIEW: The international mouse phenotyping consortium (IMPC) is producing defined gene knockout mouse lines. Here, a phenotyping program is presented that is based on micro-computed tomography (µCT) assessment of distal femur and vertebra. Lines with significant variation undergo a computer-based bone histomorphometric analysis. RECENT FINDINGS: Of the 220 lines examined to date, approximately 15% have a significant variation (high or low) by µCT, most of which are not identified by the IMPC screen. Significant dimorphism between the sexes and bone compartments adds to the complexity of the skeletal findings. The µCT information that is posted at www.bonebase.org can group KOMP lines with similar morphological features. The histological data is presented in a graphic form that associates the cellular features with a specific anatomic group. The web portal presents a bone-centric view appropriate for the skeletal biologist/clinician to organize and understand the large number of genes that can influence skeletal health. Cataloging the relative severity of each variant is the first step towards compiling the dataset necessary to appreciate the full polygenic basis of degenerative bone disease.


Subject(s)
Bone and Bones/diagnostic imaging , Femur/diagnostic imaging , Spine/diagnostic imaging , Animals , Bone and Bones/pathology , Databases, Factual , Femur/pathology , Genotype , Information Management , Mice , Mice, Knockout , Phenotype , Program Development , Severity of Illness Index , Sex Characteristics , Spine/pathology , X-Ray Microtomography
18.
Int J Biol Macromol ; 106: 1159-1165, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28847606

ABSTRACT

The study aimed to investigate the localized osteogenic activity of recombinant human bone morphogenetic protein (rhBMP-2), when delivered using enzymatically crosslinkable injectable glycol chitosan hydrogel. A critical sized bilateral calvarial defect model was used wherein one defect was implanted with rhBMP-2 loaded hydrogel (HPP-GC+BMP). The neighboring defect was implanted with an osteoconductive, collagen hydroxyapatite matrix "Healos®". The implantation of HPP-GC+BMP led to complete closure of the critical sized defect at 4 weeks post-implantation. The neighboring site implanted with Healos® however, did not show any bone formation. The spatial control of rhBMP-2 bioactivity at the cellular level was confirmed by histological and histomorphometric analysis of the calvaria isolated from Col3.6 transgenic animals which can express fluorescence in osteoblast and pre-osteoblast cells. The retained rhBMP-2 in HPPGC+BMP implant was able to localize osteoprogenitor recruitment and osteogenesis, at the implantation site. The results demonstrate the efficacy of HPP-GC hydrogel in minimizing the diffusive loss of rhBMP-2 from the implantation site, compared to the collagen hydroxyapatite scaffold.


Subject(s)
Bone Morphogenetic Protein 2/administration & dosage , Osteogenesis/drug effects , Recombinant Proteins/administration & dosage , Skull/growth & development , Animals , Bone Morphogenetic Protein 2/chemistry , Bone Morphogenetic Protein 2/genetics , Bone-Implant Interface/growth & development , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/administration & dosage , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Mice , Mice, Transgenic , Recombinant Proteins/genetics , Skull/drug effects
19.
Dev Biol ; 426(1): 56-68, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28438606

ABSTRACT

Limb synovial joints are composed of distinct tissues, but it is unclear which progenitors produce those tissues and how articular cartilage acquires its functional postnatal organization characterized by chondrocyte columns, zone-specific cell volumes and anisotropic matrix. Using novel Gdf5CreERT2 (Gdf5-CE), Prg4-CE and Dkk3-CE mice mated to R26-Confetti or single-color reporters, we found that knee joint progenitors produced small non-migratory progenies and distinct local tissues over prenatal and postnatal time. Stereological imaging and quantification indicated that the columns present in juvenile-adult tibial articular cartilage consisted of non-daughter, partially overlapping lineage cells, likely reflecting cell rearrangement and stacking. Zone-specific increases in cell volume were major drivers of tissue thickening, while cell proliferation or death played minor roles. Second harmonic generation with 2-photon microscopy showed that the collagen matrix went from being isotropic and scattered at young stages to being anisotropic and aligned along the cell stacks in adults. Progenitor tracing at prenatal or juvenile stages showed that joint injury provoked a massive and rapid increase in synovial Prg4+ and CD44+/P75+ cells some of which filling the injury site, while neighboring chondrocytes appeared unresponsive. Our data indicate that local cell populations produce distinct joint tissues and that articular cartilage growth and zonal organization are mainly brought about by cell volume expansion and topographical cell rearrangement. Synovial Prg4+ lineage progenitors are exquisitely responsive to acute injury and may represent pioneers in joint tissue repair.


Subject(s)
Cartilage, Articular , Cell Size , Chondrogenesis/physiology , Knee Injuries/metabolism , Knee Joint/growth & development , Mesenchymal Stem Cells/metabolism , Animals , Cartilage, Articular/cytology , Cartilage, Articular/embryology , Cartilage, Articular/growth & development , Cartilage, Articular/injuries , Cell Differentiation/physiology , Cell Lineage , Cell Proliferation , Chondrocytes/cytology , Collagen/metabolism , Growth Differentiation Factor 5/metabolism , Knee Joint/cytology , Mice , Mice, Transgenic , Synovial Membrane/cytology
20.
Oncotarget ; 7(51): 83837-83842, 2016 Dec 20.
Article in English | MEDLINE | ID: mdl-27863377

ABSTRACT

Generalized arterial calcification of infancy (GACI) is an autosomal recessive disorder characterized by early onset of extensive mineralization of the cardiovascular system. The classical forms of GACI are caused by mutations in the ENPP1 gene, encoding a membrane-bound pyrophosphatase/phosphodiesterase that hydrolyzes ATP to AMP and inorganic pyrophosphate. The asj-2J mouse harboring a spontaneous mutation in the Enpp1 gene has been characterized as a model for GACI. These mutant mice develop ectopic mineralization in skin and vascular connective tissues as well as in cartilage and collagen-rich tendons and ligaments. This study examined in detail the temporal ectopic mineralization phenotype of connective tissues in this mouse model, utilizing a novel cryo-histological method that does not require decalcification of bones. The wild type, heterozygous, and homozygous mice were administered fluorescent mineralization labels at 4 weeks (calcein), 10 weeks (alizarin complexone), and 11 weeks of age (demeclocycline). Twenty-four hours later, outer ears, muzzle skin, trachea, aorta, shoulders, and vertebrae were collected from these mice and examined for progression of mineralization. The results revealed differential timeline for disease initiation and progression in various tissues of this mouse model. It also highlights the advantages of cryo-histological fluorescent imaging technique to study mineral deposition in mouse models of ectopic mineralization disorders.


Subject(s)
Connective Tissue/pathology , Mutation , Phosphoric Diester Hydrolases/genetics , Pyrophosphatases/genetics , Vascular Calcification/genetics , Vascular Calcification/pathology , Animals , Anthraquinones/administration & dosage , Connective Tissue/enzymology , Demeclocycline/administration & dosage , Disease Progression , Fluoresceins/administration & dosage , Fluorescent Dyes/administration & dosage , Genetic Predisposition to Disease , Heterozygote , Homozygote , Mice, Inbred BALB C , Mice, Mutant Strains , Microscopy, Fluorescence/methods , Phenotype , Phosphoric Diester Hydrolases/metabolism , Pyrophosphatases/metabolism , Time Factors , Vascular Calcification/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...