62 results on '"Carolyn A. Meyers"'
Search Results
2. Interaction between the nervous and skeletal systems
- Author
-
Jiajia Xu, Zhongmin Zhang, Junjie Zhao, Carolyn A. Meyers, Seungyong Lee, Qizhi Qin, and Aaron W. James
- Subjects
nervous system ,skeletal system ,crosstalk ,bone repair and regeneration ,nerve innervation ,Biology (General) ,QH301-705.5 - Abstract
The skeleton is one of the largest organ systems in the body and is richly innervated by the network of nerves. Peripheral nerves in the skeleton include sensory and sympathetic nerves. Crosstalk between bones and nerves is a hot topic of current research, yet it is not well understood. In this review, we will explore the role of nerves in bone repair and remodeling, as well as summarize the molecular mechanisms by which neurotransmitters regulate osteogenic differentiation. Furthermore, we discuss the skeleton’s role as an endocrine organ that regulates the innervation and function of nerves by secreting bone-derived factors. An understanding of the interactions between nerves and bone can help to prevent and treat bone diseases caused by abnormal innervation or nerve function, develop new strategies for clinical bone regeneration, and improve patient outcomes.
- Published
- 2022
- Full Text
- View/download PDF
3. Systemic DKK1 neutralization enhances human adipose‐derived stem cell mediated bone repair
- Author
-
Stefano Negri, Yiyun Wang, Takashi Sono, Qizhi Qin, Ginny Ching‐Yun Hsu, Masnsen Cherief, Jiajia Xu, Seungyong Lee, Robert J. Tower, Victoria Yu, Abhi Piplani, Carolyn A. Meyers, Kristen Broderick, Min Lee, and Aaron W. James
- Subjects
adipose stem cell ,adipose stromal cell ,bone healing ,bone repair ,bone tissue engineering ,mesenchymal stem cell ,Medicine (General) ,R5-920 ,Cytology ,QH573-671 - Abstract
Abstract Progenitor cells from adipose tissue are able to induce bone repair; however, inconsistent or unreliable efficacy has been reported across preclinical and clinical studies. Soluble inhibitory factors, such as the secreted Wnt signaling antagonists Dickkopf‐1 (DKK1), are expressed to variable degrees in human adipose‐derived stem cells (ASCs), and may represent a targetable “molecular brake” on ASC mediated bone repair. Here, anti‐DKK1 neutralizing antibodies were observed to increase the osteogenic differentiation of human ASCs in vitro, accompanied by increased canonical Wnt signaling. Human ASCs were next engrafted into a femoral segmental bone defect in NOD‐Scid mice, with animals subsequently treated with systemic anti‐DKK1 or isotype control during the repair process. Human ASCs alone induced significant but modest bone repair. However, systemic anti‐DKK1 induced an increase in human ASC engraftment and survival, an increase in vascular ingrowth, and ultimately improved bone repair outcomes. In summary, anti‐DKK1 can be used as a method to augment cell‐mediated bone regeneration, and could be particularly valuable in the contexts of impaired bone healing such as osteoporotic bone repair.
- Published
- 2021
- Full Text
- View/download PDF
4. 4: Peripheral Nerves Engage in Reciprocal Neuro- and Angiogenic Crosstalk With SMCs in Extremity Trauma
- Author
-
Charles D. Hwang, MD, Chase A. Pagani, BS, Seungyong Lee, PhD, Qizhi Qin, PhD, Simone Marini, PhD, Amanda Huber, PhD, Carolyn A. Meyers, BS, Geoffrey E. Hespe, MD, Amy L. Strong, MD, PhD, David M. Stepien, MD, PhD, Michael Sorkin, MD, Johanna Nunez, MD, Aaron W. James, MD, PhD, and Benjamin Levi, MD
- Subjects
Surgery ,RD1-811 - Abstract
Purpose: Existing literature describes the interdependence between neurotrophic and vascular signals in the central nervous system. We hypothesize a similar crosstalk important to extremity healing involving the peripheral nervous system and angiogenic cells. Nerves are difficult to capture via axons found in the periphery alone. Thus, we have interrogated from publicly available single-nuclei transcriptomic data of peripheral nerve soma (dorsal root ganglia), injured by physical transection or chemically induced pain. We present a combined analysis of extremity polytrauma (burn/tenotomy HO model) and peripheral nerve (post-injury/pain DRG model) to determine if there is expression of vascular signals by nerves and reciprocal neurotrophic signals by cells local to the injury site. Methods: A 30% dorsal burn and Achilles transection was performed in C57/BL6J mice. The tendon site tissues were harvested from baseline (t0) and day 7, 42 after induction. Samples were prepared for library generation on a 10x Genomics Chromium Controller, sequenced on a Illumina HiSeq 4000, and analyzed with Cell Ranger Software for pre-processing and alignment to the mm10 genome. DRG analyses and clusters were abstracted from NIH-GEO (GSE154659). Downstream analyses including unsupervised clustering downstream analyses were performed with Seurat. Results: We first examined candidate neurotrophins and vascular signals in nerve (DRG), finding robust upregulation of Bdnf and Vegfa. In HO, the site of injury contains many cells that may potentially respond to these signals. Indeed, in sequencing data from the pre-HO anlagen, endothelium and smooth muscle cell populations express upregulation for receptors to the nerve-derived Vegfa via Flt1/VEGFR1. This population in addition to being sensitive to the VEGFA ligand, also demonstrates upregulation of Ngf, signifying a potential vasculo-neuro axis where a vascular signal induces endothelium/SMCs to produce neurotrophic signals. Completing the circuit, the original DRG cells and by logical extension, regenerating peripheral nerves, are highly enriched for the neurotrophin receptors: Ntrk1/TrkA (responsive to the SMC derived NGF), Ntrk2/TrkB (responsive to the nerve-autonomous BDNF), and Ntrk3/TrkC (partial combined NGF/BDNF response). This potentially signifies a feedforward loop where peripheral nerve induces angiogenesis which in return, promotes nascent nerve ingrowth in a cyclical process. Indeed, in targeted knockout of a local VEGFA source (VegfaPrrx1 mice), the injury site demonstrates parallel reduction in vascular density (77%) and reduction in nerve fiber frequency (62%) within the HO site. Conclusions: These findings represent the first work characterizing the coordination between neurogenic and angiogenic transcription programs following extremity trauma. We demonstrate through NextGen sequencing, evidence of neuroangiogenic crosstalk following musculoskeletal/neural injury. This VEGFA/NGF axis involves vascular signaling as a potential source for additional proliferation of NGF expressing pericyte/SMCs. The presented data describe the potential nerve-driven regulation contributing to the formation of HO at the extremity that with antagonism or inhibition may lead to better treatments for aberrant extremity wound healing.
- Published
- 2021
- Full Text
- View/download PDF
5. A Neurotrophic Mechanism Directs Sensory Nerve Transit in Cranial Bone
- Author
-
Carolyn A. Meyers, Seungyong Lee, Takashi Sono, Jiajia Xu, Stefano Negri, Ye Tian, Yiyun Wang, Zhu Li, Sarah Miller, Leslie Chang, Yongxing Gao, Liliana Minichiello, Thomas L. Clemens, and Aaron W. James
- Subjects
NGF ,TrkA ,osteogenesis ,bone healing ,calvarial bone ,Biology (General) ,QH301-705.5 - Abstract
Summary: The flat bones of the skull are densely innervated during development, but little is known regarding their role during repair. We describe a neurotrophic mechanism that directs sensory nerve transit in the mouse calvaria. Patent cranial suture mesenchyme represents an NGF (nerve growth factor)-rich domain, in which sensory nerves transit. Experimental calvarial injury upregulates Ngf in an IL-1β/TNF-α-rich defect niche, with consequent axonal ingrowth. In calvarial osteoblasts, IL-1β and TNF-α stimulate Ngf and downstream NF-κB signaling. Locoregional deletion of Ngf delays defect site re-innervation and blunted repair. Genetic disruption of Ngf among LysM-expressing macrophages phenocopies these observations, whereas conditional knockout of Ngf among Pdgfra-expressing cells does not. Finally, inhibition of TrkA catalytic activity similarly delays re-innervation and repair. These results demonstrate an essential role of NGF-TrkA signaling in bone healing and implicate macrophage-derived NGF-induced ingrowth of skeletal sensory nerves as an important mediator of this repair.
- Published
- 2020
- Full Text
- View/download PDF
6. WISP-1 drives bone formation at the expense of fat formation in human perivascular stem cells
- Author
-
Carolyn A. Meyers, Jiajia Xu, Greg Asatrian, Catherine Ding, Jia Shen, Kristen Broderick, Kang Ting, Chia Soo, Bruno Peault, and Aaron W. James
- Subjects
Perivascular Niche ,FACS Purification ,Segmental Spinal Fusion ,Adipogenic Differentiation Conditions ,Osteogenic Differentiation ,Medicine ,Science - Abstract
Abstract The vascular wall within adipose tissue is a source of mesenchymal progenitors, referred to as perivascular stem/stromal cells (PSC). PSC are isolated via fluorescence activated cell sorting (FACS), and defined as a bipartite population of pericytes and adventitial progenitor cells (APCs). Those factors that promote the differentiation of PSC into bone or fat cell types are not well understood. Here, we observed high expression of WISP-1 among human PSC in vivo, after purification, and upon transplantation in a bone defect. Next, modulation of WISP-1 expression was performed, using WISP-1 overexpression, WISP-1 protein, or WISP-1 siRNA. Results demonstrated that WISP-1 is expressed in the perivascular niche, and high expression is maintained after purification of PSC, and upon transplantation in a bone microenvironment. In vitro studies demonstrate that WISP-1 has pro-osteogenic/anti-adipocytic effects in human PSC, and that regulation of BMP signaling activity may underlie these effects. In summary, our results demonstrate the importance of the matricellular protein WISP-1 in regulation of the differentiation of human stem cell types within the perivascular niche. WISP-1 signaling upregulation may be of future benefit in cell therapy mediated bone tissue engineering, for the healing of bone defects or other orthopaedic applications.
- Published
- 2018
- Full Text
- View/download PDF
7. Abstract 104: Identification of the Role of Sensory Nerve TrkA Signaling on Progenitor Cell Fate after Extremity Trauma
- Author
-
Charles Hwang, BS, David M. Stepien, MD, PhD, Carrie Kubiak, MD, Carolyn A. Meyers, BS, Seungyong Lee, PhD, Michael Sorkin, MD, Chase A. Pagani, BA, Talis Rehse, BS, Noelle D. Visser, MS, Mohamed A. Garada, Zaid N. Khatib, BS, Prasanth Kotha, Jeffrey Lisiecki, MD, Kaetlin Vasquez, MS, Paul S. Cederna, MD, Stephen W.P. Kemp, PhD, Thomas L. Clemens, PhD, Aaron W. James, MD, PhD, and Benjamin Levi, MD
- Subjects
Surgery ,RD1-811 - Published
- 2019
- Full Text
- View/download PDF
8. Human perivascular stem cells prevent bone graft resorption in osteoporotic contexts by inhibiting osteoclast formation
- Author
-
Stefano Negri, Kristen P. Broderick, Ginny Ching Yun Hsu, Aaron W. James, Yiyun Wang, Jiajia Xu, Qizhi Qin, Takashi Sono, Seungyong Lee, Bruno Péault, Carolyn A. Meyers, and Kenneth W. Witwer
- Subjects
musculoskeletal diseases ,0301 basic medicine ,Stromal cell ,Nude ,bone graft ,Osteoclasts ,Bone resorption ,Rats, Nude ,03 medical and health sciences ,Paracrine signalling ,0302 clinical medicine ,Osteoclast ,Tissue Engineering and Regenerative Medicine ,pericyte ,perivascular stem cell ,medicine ,Animals ,Humans ,Bone Resorption ,Bone regeneration ,mesenchymal stem cell ,spine fusion ,Chemistry ,Stem Cells ,Mesenchymal stem cell ,adipose stem cell ,Osteoblast ,Cell Biology ,General Medicine ,Rats ,Cell biology ,Resorption ,030104 developmental biology ,medicine.anatomical_structure ,osteoclast ,Osteoporosis ,Female ,Transcriptome ,030217 neurology & neurosurgery ,Stem Cell Transplantation ,Developmental Biology - Abstract
The vascular wall stores mesenchymal progenitor cells which are able to induce bone regeneration, via direct and paracrine mechanisms. Although much is known regarding perivascular cell regulation of osteoblasts, their regulation of osteoclasts, and by extension utility in states of high bone resorption, is not known. Here, human perivascular stem cells (PSCs) were used as a means to prevent autograft resorption in a gonadectomy‐induced osteoporotic spine fusion model. Furthermore, the paracrine regulation by PSCs of osteoclast formation was evaluated, using coculture, conditioned medium, and purified extracellular vesicles. Results showed that PSCs when mixed with autograft bone induce an increase in osteoblast:osteoclast ratio, promote bone matrix formation, and prevent bone graft resorption. The confluence of these factors resulted in high rates of fusion in an ovariectomized rat lumbar spine fusion model. Application of PSCs was superior across metrics to either the use of unpurified, culture‐defined adipose‐derived stromal cells or autograft bone alone. Under coculture conditions, PSCs negatively regulated osteoclast formation and did so via secreted, nonvesicular paracrine factors. Total RNA sequencing identified secreted factors overexpressed by PSCs which may explain their negative regulation of graft resorption. In summary, PSCs reduce osteoclast formation and prevent bone graft resorption in high turnover states such as gonadectomy‐induced osteoporosis., Perivascular stem cells (PSCs) derived from adipose tissue prevent bone graft resorption and enhance arthrodesis in a preclinical osteoporotic spinal fusion model. PSCs inhibit osteoclasts formation via paracrine secretion of negative regulators in a high bone turnover environment.
- Published
- 2020
- Full Text
- View/download PDF
9. NGF-p75 signaling coordinates skeletal cell migration during bone repair
- Author
-
Jiajia Xu, Zhao Li, Robert J. Tower, Stefano Negri, Yiyun Wang, Carolyn A. Meyers, Takashi Sono, Qizhi Qin, Amy Lu, Xin Xing, Edward F. McCarthy, Thomas L. Clemens, and Aaron W. James
- Subjects
Mice ,Multidisciplinary ,Osteoblasts ,Cell Movement ,Osteogenesis ,Nerve Growth Factor ,Receptors ,Animals ,Receptors, Nerve Growth Factor ,Signal Transduction - Abstract
Bone regeneration following injury is initiated by inflammatory signals and occurs in association with infiltration by sensory nerve fibers. Together, these events are believed to coordinate angiogenesis and tissue reprogramming, but the mechanism of coupling immune signals to reinnervation and osteogenesis is unknown. Here, we found that nerve growth factor (NGF) is expressed following cranial bone injury and signals via p75 in resident mesenchymal osteogenic precursors to affect their migration into the damaged tissue. Mice lacking Ngf in myeloid cells demonstrated reduced migration of osteogenic precursors to the injury site with consequently delayed bone healing. These features were phenocopied by mice lacking p75 in Pdgfra + osteoblast precursors. Single-cell transcriptomics identified mesenchymal subpopulations with potential roles in cell migration and immune response, altered in the context of p75 deletion. Together, these results identify the role of p75 signaling pathway in coordinating skeletal cell migration during early bone repair.
- Published
- 2022
10. NGF-p75 signaling coordinates skeletal cell migration during bone repair
- Author
-
Takashi Sono, Zhu Li, Jing Xu, Qizhi Qin, Robert J. Tower, Thomas L. Clemens, Aaron W. James, Carolyn A. Meyers, Amy Lu, Stefano Negri, Yuefan Wang, and Edward F. McCarthy
- Subjects
medicine.anatomical_structure ,Nerve growth factor ,biology ,Precursor cell ,Mesenchymal stem cell ,medicine ,biology.protein ,Cell migration ,Progenitor cell ,Wound healing ,Sensory nerve ,Cell biology ,Neurotrophin - Abstract
Bone regeneration following injury is initiated by inflammatory signals and occurs in association with infiltration by sensory nerve fibers. Together, these events are believed to coordinate angiogenesis and tissue reprogramming, but the mechanism of coupling immune signals to re-innervation and osteogenesis is unknown. Here, we found that NGF is expressed following cranial bone injury and signals via p75 in resident mesenchymal osteogenic precursors to impact their migration into the damaged tissue. Mice lacking Ngf in myeloid cells demonstrated reduced migration of osteogenic precursors to the injury site with consequently delayed bone healing. These features were phenocopied by mice lacking p75 in Pdgfra+ osteoblast precursors. Single-cell transcriptomics identified mesenchymal subpopulations with potential roles in cell migration and immune response, altered in the context of p75 deletion. Together, these results identify the role of p75 signaling pathway in coordinating skeletal cell migration during early bone repair.
- Published
- 2021
- Full Text
- View/download PDF
11. 4: Peripheral Nerves Engage in Reciprocal Neuro- and Angiogenic Crosstalk With SMCs in Extremity Trauma
- Author
-
Amanda K. Huber, Aaron W. James, David M. Stepien, Geoffrey E. Hespe, Simone Marini, Amy L. Strong, Seungyong Lee, Johanna Nunez, Benjamin Levi, Qizhi Qin, Michael Sorkin, Chase A. Pagani, Charles Hwang, and Carolyn A. Meyers
- Subjects
Crosstalk (biology) ,RD1-811 ,nervous system ,business.industry ,PSRC 2021 Abstract Supplement ,Medicine ,Surgery ,business ,Neuroscience ,Reciprocal ,Peripheral - Abstract
Purpose: Existing literature describes the interdependence between neurotrophic and vascular signals in the central nervous system. We hypothesize a similar crosstalk important to extremity healing involving the peripheral nervous system and angiogenic cells. Nerves are difficult to capture via axons found in the periphery alone. Thus, we have interrogated from publicly available single-nuclei transcriptomic data of peripheral nerve soma (dorsal root ganglia), injured by physical transection or chemically induced pain. We present a combined analysis of extremity polytrauma (burn/tenotomy HO model) and peripheral nerve (post-injury/pain DRG model) to determine if there is expression of vascular signals by nerves and reciprocal neurotrophic signals by cells local to the injury site. Methods: A 30% dorsal burn and Achilles transection was performed in C57/BL6J mice. The tendon site tissues were harvested from baseline (t0) and day 7, 42 after induction. Samples were prepared for library generation on a 10x Genomics Chromium Controller, sequenced on a Illumina HiSeq 4000, and analyzed with Cell Ranger Software for pre-processing and alignment to the mm10 genome. DRG analyses and clusters were abstracted from NIH-GEO (GSE154659). Downstream analyses including unsupervised clustering downstream analyses were performed with Seurat. Results: We first examined candidate neurotrophins and vascular signals in nerve (DRG), finding robust upregulation of Bdnf and Vegfa. In HO, the site of injury contains many cells that may potentially respond to these signals. Indeed, in sequencing data from the pre-HO anlagen, endothelium and smooth muscle cell populations express upregulation for receptors to the nerve-derived Vegfa via Flt1/VEGFR1. This population in addition to being sensitive to the VEGFA ligand, also demonstrates upregulation of Ngf, signifying a potential vasculo-neuro axis where a vascular signal induces endothelium/SMCs to produce neurotrophic signals. Completing the circuit, the original DRG cells and by logical extension, regenerating peripheral nerves, are highly enriched for the neurotrophin receptors: Ntrk1/TrkA (responsive to the SMC derived NGF), Ntrk2/TrkB (responsive to the nerve-autonomous BDNF), and Ntrk3/TrkC (partial combined NGF/BDNF response). This potentially signifies a feedforward loop where peripheral nerve induces angiogenesis which in return, promotes nascent nerve ingrowth in a cyclical process. Indeed, in targeted knockout of a local VEGFA source (VegfaPrrx1 mice), the injury site demonstrates parallel reduction in vascular density (77%) and reduction in nerve fiber frequency (62%) within the HO site. Conclusions: These findings represent the first work characterizing the coordination between neurogenic and angiogenic transcription programs following extremity trauma. We demonstrate through NextGen sequencing, evidence of neuroangiogenic crosstalk following musculoskeletal/neural injury. This VEGFA/NGF axis involves vascular signaling as a potential source for additional proliferation of NGF expressing pericyte/SMCs. The presented data describe the potential nerve-driven regulation contributing to the formation of HO at the extremity that with antagonism or inhibition may lead to better treatments for aberrant extremity wound healing.
- Published
- 2021
12. Skeletogenic Capacity of Human Perivascular Stem Cells Obtained Via Magnetic-Activated Cell Sorting
- Author
-
Greg Asatrian, Erin Zou, Aaron W. James, Leslie Chang, Leititia Zhang, Catherine Ding, Bruno Péault, Min Lee, Noah Yan, Kristen P. Broderick, Jiajia Xu, Yiyun Wang, and Carolyn A. Meyers
- Subjects
Adult ,Stromal cell ,medicine.medical_treatment ,0206 medical engineering ,Population ,Biomedical Engineering ,Antigens, CD34 ,Bioengineering ,Cell Separation ,02 engineering and technology ,Biology ,Biochemistry ,Biomaterials ,03 medical and health sciences ,Osteogenesis ,medicine ,Humans ,Cell Lineage ,education ,030304 developmental biology ,Wound Healing ,0303 health sciences ,education.field_of_study ,Magnetic-activated cell sorting ,Magnetic Phenomena ,Stem Cells ,Skull ,Mesenchymal stem cell ,Cell Differentiation ,Original Articles ,Stem-cell therapy ,Cell sorting ,020601 biomedical engineering ,Cell biology ,Adipose Tissue ,CD146 ,Stem cell ,Biomarkers - Abstract
Human perivascular stem/stromal cells (PSC) are a multipotent mesenchymal progenitor cell population defined by their perivascular residence. PSC are increasingly studied for their application in skeletal regenerative medicine. PSC from subcutaneous white adipose tissue are most commonly isolated via fluorescence-activated cell sorting (FACS), and defined as a bipartite population of CD146(+)CD34(−)CD31(−)CD45(−) pericytes and CD34(+)CD146(−)CD31(−)CD45(−) adventitial cells. FACS poses several challenges for clinical translation, including requirements for facilities, equipment, and personnel. The purpose of this study is to identify if magnetic-activated cell sorting (MACS) is a feasible method to derive PSC, and to determine if MACS-derived PSC are comparable to our previous experience with FACS-derived PSC. In brief, CD146(+) pericytes and CD34(+) adventitial cells were enriched from human lipoaspirate using a multistep column approach. Next, cell identity and purity were analyzed by flow cytometry. In vitro multilineage differentiation studies were performed with MACS-defined PSC subsets. Finally, in vivo application was performed in nonhealing calvarial bone defects in Scid mice. Results showed that human CD146(+) pericytes and CD34(+) adventitial cells may be enriched by MACS, with defined purity, anticipated cell surface marker expression, and capacity for multilineage differentiation. In vivo, MACS-derived PSC induce ossification of bone defects. These data document the feasibility of a MACS approach for the enrichment and application of PSC in the field of tissue engineering and regenerative medicine. IMPACT STATEMENT: Our findings suggest that perivascular stem/stromal cells, and in particular adventitial cells, may be isolated by magnetic-activated cell sorting and applied as an uncultured autologous stem cell therapy in a same-day setting for bone defect repair.
- Published
- 2019
- Full Text
- View/download PDF
13. PDGFRα marks distinct perivascular populations with different osteogenic potential within adipose tissue
- Author
-
Ye Tian, Aaron W. James, Yiyun Wang, Jiajia Xu, Carolyn A. Meyers, Kristen P. Broderick, Yongxing Gao, and Bruno Péault
- Subjects
Male ,0301 basic medicine ,Receptor, Platelet-Derived Growth Factor alpha ,Stromal cell ,CD34 ,Adipose tissue ,Biology ,Bone morphogenetic protein 2 ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Osteogenesis ,medicine ,Animals ,Humans ,Progenitor cell ,Mesenchymal stem cell ,Cell Differentiation ,Osteoblast ,Cell Biology ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Adipose Tissue ,Molecular Medicine ,Pericyte ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
The perivascular niche within adipose tissue is known to house multipotent cells, including osteoblast precursors. However, the identity of perivascular subpopulations that may mineralize or ossify most readily is not known. Here, we utilize inducible PDGFRα (platelet-derived growth factor alpha) reporter animals to identify subpopulations of perivascular progenitor cells. Results showed that PDGFRα-expressing cells are present in four histologic niches within inguinal fat, including two perivascular locations. PDGFRα+ cells are most frequent within the tunica adventitia of arteries and veins, where PDGFRα+ cells populate the inner aspects of the adventitial layer. Although both PDGFRα+ and PDGFRα− fractions are multipotent progenitor cells, adipose tissue-derived PDGFRα+ stromal cells proliferate faster and mineralize to a greater degree than their PDGFRα− counterparts. Likewise, PDGFRα+ ectopic implants reconstitute the perivascular niche and ossify to a greater degree than PDGFRα− cell fractions. Adventicytes can be further grouped into three distinct groups based on expression of PDGFRα and/or CD34. When further partitioned, adventicytes co-expressing PDGFRα and CD34 represented a cell fraction with the highest mineralization potential. Long-term tracing studies showed that PDGFRα-expressing adventicytes give rise to adipocytes, but not to other cells within the vessel wall under homeostatic conditions. However, upon bone morphogenetic protein 2 (BMP2)-induced ossicle formation, descendants of PDGFRα+ cells gave rise to osteoblasts, adipocytes, and “pericyte-like” cells within the ossicle. In sum, PDGFRα marks distinct perivascular osteoprogenitor cell subpopulations within adipose tissue. The identification of perivascular osteoprogenitors may contribute to our improved understanding of pathologic mineralization/ossification.
- Published
- 2019
- Full Text
- View/download PDF
14. Comparison of Human Tissue Microarray to Human Pericyte Transcriptome Yields Novel Perivascular Cell Markers
- Author
-
Sarah Miller, Aaron W. James, Winters R. Hardy, Catherine Ding, Ching Yun Hsu, Bruno Péault, Carolyn A. Meyers, and Mario Gomez Salazar
- Subjects
0301 basic medicine ,Proteome ,Biology ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Original Research Reports ,Antigens, CD ,medicine ,Humans ,Perivascular Cell ,Progenitor cell ,Cells, Cultured ,Tissue microarray ,Gene Expression Profiling ,Mesenchymal stem cell ,Cell Biology ,Hematology ,Flow Cytometry ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Adipose Tissue ,Tissue Array Analysis ,Pericyte ,Pericytes ,Software ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Human perivascular progenitor cells, including pericytes, are well-described multipotent mesenchymal cells giving rise to mesenchymal stem cells in culture. Despite the unique location of pericytes, specific antigens to distinguish human pericytes from other cell types are few. Here, we employed a human tissue microarray (Human Protein Atlas) to identify proteins that are strongly and specifically expressed in a pericytic location within human adipose tissue. Next, these results were cross-referenced with RNA sequencing data from human adipose tissue pericytes, as defined as a fluorescence activated cell sorting (FACS) purified CD146(+)CD34(−)CD31(−)CD45(−) cell population. Results showed that from 105,532 core biopsies of soft tissue, 229 proteins showed strong and specific perivascular immunoreactivity, the majority of which (155) were present in the tunica intima. Next, cross-referencing with the transcriptome of FACS-derived CD146(+) pericytes yielded 25 consistently expressed genes/proteins, including 18 novel antigens. A majority of these transcripts showed maintained expression after culture propagation (56% of genes). Interestingly, many novel antigens within pericytes are regulators of osteogenic differentiation. In sum, our study demonstrates the existence of novel pericyte markers, some of which are conserved in culture that may be useful for future efforts to typify, isolate, and characterize human pericytes.
- Published
- 2019
- Full Text
- View/download PDF
15. Assessing the Bone-Forming Potential of Pericytes
- Author
-
Carolyn A, Meyers, Chenchao, Wang, Jiajia, Xu, Hsin Chuan, Pan, Jia, Shen, Kang, Ting, Chia, Soo, Bruno M, Péault, and Aaron W, James
- Subjects
Bone Regeneration ,Tissue Engineering ,Bone Marrow Cells ,Cell Differentiation ,Mesenchymal Stem Cells ,Cell Separation ,Cell Line ,Rats ,Mice ,Adipose Tissue ,Osteogenesis ,Animals ,Humans ,Pericytes - Abstract
Human pericytes are a perivascular cell population with mesenchymal stem cell properties, present in all vascularized tissues. Human pericytes have a distinct immunoprofile, which may be leveraged for purposes of cell purification. Adipose tissue is the most commonly used cell source for human pericyte derivation. Pericytes can be isolated by FACS (fluorescence-activated cell sorting), most commonly procured from liposuction aspirates. Pericytes have clonal multilineage differentiation potential, and their potential utility for bone regeneration has been described across multiple animal models. The following review will discuss in vivo methods for assessing the bone-forming potential of purified pericytes. Potential models include (1) mouse intramuscular implantation, (2) mouse calvarial defect implantation, and (3) rat spinal fusion models. In addition, the presented surgical protocols may be used for the in vivo analysis of other osteoprogenitor cell types.
- Published
- 2021
16. Assessing the Bone-Forming Potential of Pericytes
- Author
-
Hsin Chuan Pan, Jiajia Xu, Kang Ting, Carolyn A. Meyers, Jia Shen, Bruno Péault, Aaron W. James, Chenchao Wang, and Chia Soo
- Subjects
0301 basic medicine ,education.field_of_study ,Cell type ,Mesenchymal stem cell ,Population ,Biology ,Cell sorting ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Tissue engineering ,medicine ,Pericyte ,Stem cell ,Bone regeneration ,education ,030217 neurology & neurosurgery - Abstract
Human pericytes are a perivascular cell population with mesenchymal stem cell properties, present in all vascularized tissues. Human pericytes have a distinct immunoprofile, which may be leveraged for purposes of cell purification. Adipose tissue is the most commonly used cell source for human pericyte derivation. Pericytes can be isolated by FACS (fluorescence-activated cell sorting), most commonly procured from liposuction aspirates. Pericytes have clonal multilineage differentiation potential, and their potential utility for bone regeneration has been described across multiple animal models. The following review will discuss in vivo methods for assessing the bone-forming potential of purified pericytes. Potential models include (1) mouse intramuscular implantation, (2) mouse calvarial defect implantation, and (3) rat spinal fusion models. In addition, the presented surgical protocols may be used for the in vivo analysis of other osteoprogenitor cell types.
- Published
- 2021
- Full Text
- View/download PDF
17. NGF-TrkA signaling dictates neural ingrowth and aberrant osteochondral differentiation after soft tissue trauma
- Author
-
Chase A. Pagani, Stefano Negri, Amanda K. Huber, Charles Hwang, Benjamin Levi, Carolyn A. Meyers, Jiajia Xu, Robert J. Tower, Stephen W. P. Kemp, Aaron W. James, Michael Sorkin, Husain Rasheed, Qizhi Qin, Yuxiao Sun, David M. Stepien, Seungyong Lee, Sarah Miller, Carrie A. Kubiak, Thomas L. Clemens, Paul S. Cederna, Noelle D. Visser, Liliana Minichiello, Simone Marini, and Yiyun Wang
- Subjects
Cellular differentiation ,Science ,General Physics and Astronomy ,Cartilage metabolism ,Biology ,Tropomyosin receptor kinase A ,Fibroblast growth factor ,Inbred C57BL ,General Biochemistry, Genetics and Molecular Biology ,Article ,Mice ,Osteogenesis ,Nerve Growth Factor ,Animals ,Receptor, trkA ,Bone ,Denervation ,Multidisciplinary ,Stem Cells ,fungi ,Growth factor signalling ,food and beverages ,Cell Differentiation ,General Chemistry ,Axons ,Cell biology ,Mice, Inbred C57BL ,Nerve growth factor ,Cartilage ,nervous system ,trkA ,Wounds and Injuries ,Signal transduction ,Stem cell ,Peripheral nervous system ,Signal Transduction ,Receptor - Abstract
Pain is a central feature of soft tissue trauma, which under certain contexts, results in aberrant osteochondral differentiation of tissue-specific stem cells. Here, the role of sensory nerve fibers in this abnormal cell fate decision is investigated using a severe extremity injury model in mice. Soft tissue trauma results in NGF (Nerve growth factor) expression, particularly within perivascular cell types. Consequently, NGF-responsive axonal invasion occurs which precedes osteocartilaginous differentiation. Surgical denervation impedes axonal ingrowth, with significant delays in cartilage and bone formation. Likewise, either deletion of Ngf or two complementary methods to inhibit its receptor TrkA (Tropomyosin receptor kinase A) lead to similar delays in axonal invasion and osteochondral differentiation. Mechanistically, single-cell sequencing suggests a shift from TGFβ to FGF signaling activation among pre-chondrogenic cells after denervation. Finally, analysis of human pathologic specimens and databases confirms the relevance of NGF-TrkA signaling in human disease. In sum, NGF-mediated TrkA-expressing axonal ingrowth drives abnormal osteochondral differentiation after soft tissue trauma. NGF-TrkA signaling inhibition may have dual therapeutic use in soft tissue trauma, both as an analgesic and negative regulator of aberrant stem cell differentiation., Soft tissue trauma can result in aberrant osteochondral differentiation of local mesenchymal progenitor cells. Here the authors show that, in mice, soft tissue trauma results in NGF expression by perivascular cells, which leads to axonal invasion and drives abnormal osteochondral differentiation, and show that this process can be prevented by inhibition of NGF signaling.
- Published
- 2021
18. Author response: Lysosomal protein surface expression discriminates fat- from bone-forming human mesenchymal precursor cells
- Author
-
Stefano Negri, Ching Yun Hsu, Yongxing Gao, Kristen P. Broderick, Leslie Chang, Yiyun Wang, Ye Tian, Nusrat Kahn, Winters R. Hardy, Carolyn A. Meyers, Robert J. Tower, Bruno Péault, Shuaishuai Hu, Aaron W. James, Takashi Sono, and Jiajia Xu
- Subjects
Chemistry ,Precursor cell ,Mesenchymal stem cell ,Bone forming ,Surface protein ,Cell biology - Published
- 2020
- Full Text
- View/download PDF
19. Endogenous CCN family member WISP1 inhibits trauma-induced heterotopic ossification
- Author
-
Sarah Miller, Jiajia Xu, Charles Hwang, Stefano Negri, Carolyn A. Meyers, Benjamin Levi, Simone Marini, David M. Stepien, Karen M. Lyons, Yiyun Wang, Ginny Ching Yun Hsu, Edward F. McCarthy, Chase A. Pagani, and Aaron W. James
- Subjects
0301 basic medicine ,Stromal cell ,1.1 Normal biological development and functioning ,Messenger ,Context (language use) ,Stem cells ,Ossification ,Biology ,CCN Intercellular Signaling Proteins ,Transcriptome ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Proto-Oncogene Proteins ,2.1 Biological and endogenous factors ,Animals ,Humans ,RNA, Messenger ,Endochondral ossification ,Neoplastic ,Ossification, Heterotopic ,Cell Differentiation ,General Medicine ,Chondrogenesis ,Cell biology ,Gene Expression Regulation, Neoplastic ,CTGF ,Cartilage ,030104 developmental biology ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,CYR61 ,RNA ,Heterotopic ,Bone Biology ,Signal transduction ,Biotechnology ,Signal Transduction ,Research Article - Abstract
Heterotopic ossification (HO) is defined as abnormal differentiation of local stromal cells of mesenchymal origin, resulting in pathologic cartilage and bone matrix deposition. Cyr61, CTGF, Nov (CCN) family members are matricellular proteins that have diverse regulatory functions on cell proliferation and differentiation, including the regulation of chondrogenesis. However, little is known regarding CCN family member expression or function in HO. Here, a combination of bulk and single-cell RNA sequencing defined the dynamic temporospatial pattern of CCN family member induction within a mouse model of trauma-induced HO. Among CCN family proteins, Wisp1 (also known as Ccn4) was most upregulated during the evolution of HO, and Wisp1 expression corresponded with chondrogenic gene profile. Immunohistochemistry confirmed WISP1 expression across traumatic and genetic HO mouse models as well as in human HO samples. Transgenic Wisp1(LacZ/LacZ) knockin animals showed an increase in endochondral ossification in HO after trauma. Finally, the transcriptome of Wisp1-null tenocytes revealed enrichment in signaling pathways, such as the STAT3 and PCP signaling pathways, that may explain increased HO in the context of Wisp1 deficiency. In sum, CCN family members, and in particular Wisp1, are spatiotemporally associated with and negatively regulate trauma-induced HO formation.
- Published
- 2020
- Full Text
- View/download PDF
20. A neurotrophic mechanism directs sensory nerve transit in cranial bone
- Author
-
Zhu Li, Ye Tian, Stefano Negri, Thomas L. Clemens, Takashi Sono, Seungyong Lee, Aaron W. James, Liliana Minichiello, Sarah Miller, Carolyn A. Meyers, Leslie Chang, Jiajia Xu, Yongxing Gao, and Yiyun Wang
- Subjects
0301 basic medicine ,Mesenchyme ,calvarial bone ,Calvaria ,Bone healing ,Biology ,Tropomyosin receptor kinase A ,General Biochemistry, Genetics and Molecular Biology ,Article ,Bone and Bones ,osteogenesis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Conditional gene knockout ,medicine ,Animals ,lcsh:QH301-705.5 ,NGF ,Animal ,TrkA ,Skull ,Cell biology ,Disease Models, Animal ,030104 developmental biology ,Nerve growth factor ,medicine.anatomical_structure ,nervous system ,lcsh:Biology (General) ,Disease Models ,biology.protein ,Bone Remodeling ,bone healing ,030217 neurology & neurosurgery ,Neurotrophin ,Sensory nerve - Abstract
SUMMARY The flat bones of the skull are densely innervated during development, but little is known regarding their role during repair. We describe a neurotrophic mechanism that directs sensory nerve transit in the mouse calvaria. Patent cranial suture mesenchyme represents an NGF (nerve growth factor)-rich domain, in which sensory nerves transit. Experimental calvarial injury upregulates Ngf in an IL-1β/TNF-α-rich defect niche, with consequent axonal ingrowth. In calvarial osteoblasts, IL-1β and TNF-α stimulate Ngf and downstream NF-κB signaling. Locoregional deletion of Ngf delays defect site re-innervation and blunted repair. Genetic disruption of Ngf among LysM-expressing macrophages phenocopies these observations, whereas conditional knockout of Ngf among Pdgfra-expressing cells does not. Finally, inhibition of TrkA catalytic activity similarly delays re-innervation and repair. These results demonstrate an essential role of NGF-TrkA signaling in bone healing and implicate macrophage-derived NGF-induced ingrowth of skeletal sensory nerves as an important mediator of this repair., In Brief Meyers et al. describe the role of skeletal sensory nerves in cranial bone repair. The authors demonstrate several necessary aspects of membranous bone healing, including influx of nerve growth factor (NGF)-expressing macrophages after injury, followed by skeletal sensory nerve ingrowth to positively regulate bone repair., Graphical Abstract
- Published
- 2020
- Full Text
- View/download PDF
21. Abstract 7: Nerve Growth Factor Derives From Pericytes And Smooth Muscle Cells After Extremity Trauma
- Author
-
Kaetlin Vasquez, Benjamin Levi, Carolyn A. Meyers, Talis Rehse, Prasanth Kotha, Carrie A. Kubiak, Amanda K. Huber, Jeffrey Lisiecki, Michael Sorkin, David M. Stepien, Noelle D. Visser, Chase A. Pagani, Aaron W. James, Joseph A. Greenstein, Seungyong Lee, Paul S. Cederna, Charles Hwang, Mohamed A. Garada, Zaid N. Khatib, Husain Rasheed, Simone Marini, and Stephen W.P. Kemp
- Subjects
Nerve growth factor ,Smooth muscle ,business.industry ,PSRC Abstract Supplement ,lcsh:Surgery ,Medicine ,Surgery ,Anatomy ,lcsh:RD1-811 ,business - Published
- 2020
22. Comparison of skeletal and soft tissue pericytes identifies CXCR4+ bone forming mural cells in human tissues
- Author
-
Jody E. Hooper, Leititia Zhang, Yongxing Gao, Leslie Chang, Sridhar Nimmagadda, Yiyun Wang, Aaron W. James, Jiajia Xu, Carol D. Morris, Dongqing Li, Ching Yun Hsu, Kristen P. Broderick, Carolyn A. Meyers, Ye Tian, Bruno Péault, and Robert J. Tower
- Subjects
0301 basic medicine ,Histology ,Stromal cell ,Physiology ,Endocrinology, Diabetes and Metabolism ,Adipose tissue ,Diseases ,lcsh:Physiology ,Mural cell ,Article ,03 medical and health sciences ,0302 clinical medicine ,Precursor cell ,medicine ,Progenitor cell ,Bone ,lcsh:QH301-705.5 ,lcsh:QP1-981 ,Chemistry ,Mesenchymal stem cell ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Biology (General) ,CD146 ,Pericyte ,030217 neurology & neurosurgery - Abstract
Human osteogenic progenitors are not precisely defined, being primarily studied as heterogeneous multipotent cell populations and termed mesenchymal stem cells (MSCs). Notably, select human pericytes can develop into bone-forming osteoblasts. Here, we sought to define the differentiation potential of CD146+ human pericytes from skeletal and soft tissue sources, with the underlying goal of defining cell surface markers that typify an osteoblastogenic pericyte. CD146+CD31−CD45− pericytes were derived by fluorescence-activated cell sorting from human periosteum, adipose, or dermal tissue. Periosteal CD146+CD31−CD45− cells retained canonical features of pericytes/MSC. Periosteal pericytes demonstrated a striking tendency to undergo osteoblastogenesis in vitro and skeletogenesis in vivo, while soft tissue pericytes did not readily. Transcriptome analysis revealed higher CXCR4 signaling among periosteal pericytes in comparison to their soft tissue counterparts, and CXCR4 chemical inhibition abrogated ectopic ossification by periosteal pericytes. Conversely, enrichment of CXCR4+ pericytes or stromal cells identified an osteoblastic/non-adipocytic precursor cell. In sum, human skeletal and soft tissue pericytes differ in their basal abilities to form bone. Diversity exists in soft tissue pericytes, however, and CXCR4+ pericytes represent an osteoblastogenic, non-adipocytic cell precursor. Indeed, enrichment for CXCR4-expressing stromal cells is a potential new tactic for skeletal tissue engineering.
- Published
- 2020
23. Platelet Derived Growth Factor Receptor-β (PDGFRβ) lineage tracing highlights perivascular cell to myofibroblast transdifferentiation during post-traumatic osteoarthritis
- Author
-
Stefano Negri, Ching Yun Hsu, Jiajia Xu, Bruno Péault, Carolyn A. Meyers, Takashi Sono, Sarah Miller, Yiyun Wang, and Aaron W. James
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Angiogenesis ,PDGFRβ ,0206 medical engineering ,Mice, Transgenic ,02 engineering and technology ,Transgenic ,Article ,Receptor, Platelet-Derived Growth Factor beta ,Experimental ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Vasculogenesis ,Growth factor receptor ,Genes, Reporter ,pericyte ,Osteoarthritis ,perivascular stem cell ,medicine ,Animals ,Orthopedics and Sports Medicine ,Cell Lineage ,Myofibroblasts ,Reporter ,DMM ,030203 arthritis & rheumatology ,biology ,Arthritis ,Transdifferentiation ,020601 biomedical engineering ,Arthritis, Experimental ,Fibrosis ,myofibroblast ,Platelet-Derived Growth Factor beta ,medicine.anatomical_structure ,Genes ,Cell Transdifferentiation ,biology.protein ,Female ,Joints ,Pericyte ,Pericytes ,Myofibroblast ,Platelet-derived growth factor receptor ,Receptor - Abstract
Pericytes ubiquitously surround capillaries and microvessels within vascularized tissues and have diverse functions after tissue injury. In addition to regulation of angiogenesis and tissue regeneration after injury, pericytes also contribute to organ fibrosis. Destabilization of the medial meniscus (DMM) phenocopies post-traumatic osteoarthritis, yet little is known regarding the impact of DMM surgery on knee joint-associated pericytes and their cellular descendants. Here, inducible platelet-derived growth factor receptor-β (PDGFRβ)-CreERT2 reporter mice were subjected to DMM surgery, and lineage tracing studies performed over an 8-week period. Results showed that at baseline PDGFRβ reporter activity highlights abluminal perivascular cells within synovial and infrapatellar fat pad (IFP) tissues. DMM induces a temporospatially patterned increase in vascular density within synovial and subsynovial tissues. Marked vasculogenesis within IFP was accompanied by expansion of PDGFRβ reporter+ perivascular cell numbers, detachment of mGFP+ descendants from vessel walls, and aberrant adoption of myofibroblastic markers among mGFP+ cells including α-SMA, ED-A, and TGF-β1. At later timepoints, fibrotic changes and vascular maturation occurred within subsynovial tissues, with the redistribution of PDGFRβ+ cellular descendants back to their perivascular niche. In sum, PDGFRβ lineage tracing allows for tracing of perivascular cell fate within the diarthrodial joint. Further, destabilization of the joint induces vascular and fibrogenic changes of the IFP accompanied by perivascular to myofibroblast transdifferentiation.
- Published
- 2020
24. Frontal Bone Healing Is Sensitive to Wnt Signaling Inhibition via Lentiviral-Encoded Beta-Catenin Short Hairpin RNA
- Author
-
Noah Yan, Carolyn A. Meyers, Chia Soo, Catherine Ding, Jiajia Xu, Lei Zhang, Aaron W. James, Shen Pang, Leslie Chang, Erin Zou, and Kang Ting
- Subjects
0301 basic medicine ,skull ,Beta-catenin ,calvaria ,Long bone ,Biomedical Engineering ,Bioengineering ,Calvaria ,Bone healing ,Biology ,Regenerative Medicine ,Biochemistry ,osteogenesis ,Biomaterials ,Small hairpin RNA ,03 medical and health sciences ,0302 clinical medicine ,medicine ,membranous bone ,Wnt signaling pathway ,Original Articles ,Materials Engineering ,bone repair ,Wnt signaling ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Musculoskeletal ,030220 oncology & carcinogenesis ,beta-catenin shRNA ,Intramembranous ossification ,biology.protein ,intramembranous bone ,Biochemistry and Cell Biology ,Bone marrow ,bone healing ,calvarial bone defect - Abstract
The Wnt/β-catenin signaling pathway plays an integral role in skeletal biology, spanning from embryonic skeletal patterning through bone maintenance and bone repair. Most experimental methods to antagonize Wnt signaling in vivo are either systemic or transient, including genetic approaches, use of small-molecule inhibitors, or neutralizing antibodies. We sought to develop a novel, localized model of prolonged Wnt/β-catenin signaling blockade by the application and validation of a lentivirus encoding β-catenin short hairpin RNA (shRNA). Efficacy of lentiviral-encoded β-catenin shRNA was first confirmed in vitro using bone marrow mesenchymal stromal cells, and in vivo using an intramedullary long bone injection model in NOD SCID mice. Next, the effects of β-catenin knockdown were assessed in a calvarial bone defect model, in which the frontal bone demonstrates enhanced bone healing associated with heightened Wnt/β-catenin signaling. Lentivirus encoding either β-catenin shRNA or random sequence shRNA with enhanced green fluorescent protein (control) was injected overlying the calvaria of NOD SCID mice and bone defects were created in either the frontal or parietal bones. Among mice treated with lentivirus encoding β-catenin shRNA, frontal bone defect healing was significantly reduced by all radiographic and histologic metrics. In contrast, parietal bone healing was minimally impacted by β-catenin shRNA. In aggregate, our data document the application and validation of a lentivirus encoding β-catenin shRNA model that represents an easily replicable tool for examining the importance of locoregional Wnt/β-catenin signaling in bone biology and regeneration.
- Published
- 2018
- Full Text
- View/download PDF
25. WISP-1 drives bone formation at the expense of fat formation in human perivascular stem cells
- Author
-
Bruno Péault, Kang Ting, Greg Asatrian, Jia Shen, Chia Soo, Aaron W. James, Catherine Ding, Jiajia Xu, Kristen P. Broderick, and Carolyn A. Meyers
- Subjects
0301 basic medicine ,Cell type ,Stromal cell ,Science ,Population ,Cell Separation ,Biology ,Bone and Bones ,Article ,Adipogenic Differentiation Conditions ,CCN Intercellular Signaling Proteins ,Fats ,03 medical and health sciences ,Osteogenesis ,Proto-Oncogene Proteins ,Humans ,Perivascular Niche ,Progenitor cell ,education ,Cells, Cultured ,Osteogenic Differentiation ,education.field_of_study ,Multidisciplinary ,Tissue Engineering ,Stem Cells ,Matricellular protein ,Mesenchymal stem cell ,Cell Differentiation ,Flow Cytometry ,FACS Purification ,Cell biology ,Up-Regulation ,Transplantation ,Segmental Spinal Fusion ,030104 developmental biology ,Adipose Tissue ,Cellular Microenvironment ,Medicine ,Stem cell ,Pericytes - Abstract
The vascular wall within adipose tissue is a source of mesenchymal progenitors, referred to as perivascular stem/stromal cells (PSC). PSC are isolated via fluorescence activated cell sorting (FACS), and defined as a bipartite population of pericytes and adventitial progenitor cells (APCs). Those factors that promote the differentiation of PSC into bone or fat cell types are not well understood. Here, we observed high expression of WISP-1 among human PSC in vivo, after purification, and upon transplantation in a bone defect. Next, modulation of WISP-1 expression was performed, using WISP-1 overexpression, WISP-1 protein, or WISP-1 siRNA. Results demonstrated that WISP-1 is expressed in the perivascular niche, and high expression is maintained after purification of PSC, and upon transplantation in a bone microenvironment. In vitro studies demonstrate that WISP-1 has pro-osteogenic/anti-adipocytic effects in human PSC, and that regulation of BMP signaling activity may underlie these effects. In summary, our results demonstrate the importance of the matricellular protein WISP-1 in regulation of the differentiation of human stem cell types within the perivascular niche. WISP-1 signaling upregulation may be of future benefit in cell therapy mediated bone tissue engineering, for the healing of bone defects or other orthopaedic applications.
- Published
- 2018
- Full Text
- View/download PDF
26. Lineage-Specific Wnt Reporter Elucidates Mesenchymal Wnt Signaling during Bone Repair
- Author
-
Lei Zhang, Zhu Li, Jiajia Xu, Noah Yan, Carolyn A. Meyers, Aaron W. James, Leslie Chang, and Erin Zou
- Subjects
Male ,0301 basic medicine ,Cell type ,Mice, Transgenic ,Context (language use) ,Bone healing ,Biology ,Article ,Pathology and Forensic Medicine ,Green fluorescent protein ,Mesoderm ,03 medical and health sciences ,0302 clinical medicine ,Genes, Reporter ,Osteogenesis ,Animals ,Enhancer ,Wnt Signaling Pathway ,Fracture Healing ,Homeodomain Proteins ,Mesenchymal stem cell ,Wnt signaling pathway ,X-Ray Microtomography ,Cell biology ,Wnt Proteins ,030104 developmental biology ,Frontal Bone ,Homeobox ,Female ,Bones of Upper Extremity ,030217 neurology & neurosurgery - Abstract
β-Catenin-dependent Wnt signaling controls numerous aspects of skeletal development and postnatal bone repair. Currently available transgenic Wnt reporter mice allow for visualization of global canonical Wnt signaling activity within skeletal tissues, without delineation of cell type. This is particularly important in a bone repair context, in which the inflammatory phase can obscure the visualization of mesenchymal cell types of interest. To tackle the issue of tissue-specific Wnt signaling, we have generated and characterized a transgenic mouse strain [termed paired related homeobox 1 (Prx1)-Wnt-green fluorescent protein (GFP), by crossing a previously validated Prx1-Cre strain with a nuclear fluorescent reporter driven by T-cell factor/lymphoid enhancer factor activity (Rosa26-Tcf/Lef-LSL-H2B-GFP)]. Prx1-Wnt-GFP animals were subject to three models of long bone and membranous bone repair (displaced forelimb fracture, tibial cortical defect, and frontal bone defect). Results showed that, irrespective of bone type, locoregional mesenchymal cell activation of Wnt signaling occurs in a defined temporospatial pattern among Prx1-Wnt-GFP mice. In summary, Prx1-Wnt-GFP reporter animals allow for improved visualization, spatial discrimination, and facile quantification of Wnt-activated mesenchymal cells within models of adult bone repair.
- Published
- 2018
- Full Text
- View/download PDF
27. WNT16 induces proliferation and osteogenic differentiation of human perivascular stem cells
- Author
-
Jia Shen, Amy Lu, Aaron W. James, and Carolyn A. Meyers
- Subjects
0301 basic medicine ,animal structures ,endocrine system diseases ,Population ,digestive system ,Article ,03 medical and health sciences ,0302 clinical medicine ,Gene expression ,Medicine ,Orthopedics and Sports Medicine ,education ,Progenitor ,education.field_of_study ,Cell growth ,business.industry ,digestive, oral, and skin physiology ,Mesenchymal stem cell ,Wnt signaling pathway ,digestive system diseases ,Cell biology ,030104 developmental biology ,Stem cell ,business ,030217 neurology & neurosurgery ,WNT3A - Abstract
Perivascular stem cells (PSC) are a progenitor population defined by their perivascular residence. Recent studies have examined the relative difference in Wnt ligands to induce PSC differentiation, including Wnt16. Here, we examine the role of Wnt16 in the proliferation and osteogenic differentiation of human PSC. Treatment of PSC with WNT16 significantly increased cell proliferation to a greater extent than did WNT3A. In addition, WNT16 showed a significant increase in osteogenic gene expression among PSC. These data demonstrate that WNT16 represents a combined mitogenic/pro-osteogenic stimulus that may play a functional role in human mesenchymal stem cell mediated bone repair.
- Published
- 2018
- Full Text
- View/download PDF
28. Bizarre parosteal osteochondromatous proliferation: 16 Cases with a focus on histologic variability
- Author
-
Edward F. McCarthy, Aaron W. James, Margaret Cocks, Carolyn A. Meyers, Elizabeth Helmke, and Laura M. Fayad
- Subjects
Osteochondroma ,Pathology ,medicine.medical_specialty ,business.industry ,Cartilage ,Cartilage tumor ,Fibrous tissue ,medicine.disease ,Article ,030218 nuclear medicine & medical imaging ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cartilaginous Tissue ,Medicine ,Orthopedics and Sports Medicine ,business - Abstract
Bizarre parosteal osteochondromatous proliferation (BPOP) is a benign bone and cartilage forming tumor occurring on the surface of bones, predominantly on the hands and feet. A defining feature of BPOP is the purplish-blue mineralization of cartilaginous tissue, known as 'blue bone.' Here, we report on an institutional series of 16 cases of BPOP, including radiographic, histologic, and histomorphometric features. All tumors were composed of some element of bone, cartilage, fibrous tissue and 'blue bone,' though the amount of each tissue sub-type varied widely. Some cases showed focal 'blue bone' only, however this was a defining feature in all cases.
- Published
- 2018
- Full Text
- View/download PDF
29. Early Immunomodulatory Effects of Implanted Human Perivascular Stromal Cells During Bone Formation
- Author
-
Jiajia Xu, Bruno Péault, Justin M. Sacks, Chia Soo, Xinli Zhang, Aaron W. James, Kristen P. Broderick, Kang Ting, Noah Yan, Greg Asatrian, Lei Zhang, Catherine Ding, Raghav Goyal, and Carolyn A. Meyers
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Stromal cell ,endocrine system diseases ,Population ,Biomedical Engineering ,Bone Matrix ,Adipose tissue ,Bioengineering ,Mice, SCID ,Biology ,Mesenchymal Stem Cell Transplantation ,digestive system ,Biochemistry ,Immunomodulation ,Biomaterials ,Mice ,03 medical and health sciences ,Osteogenesis ,medicine ,Animals ,Humans ,Bone formation ,Progenitor cell ,education ,education.field_of_study ,digestive, oral, and skin physiology ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Original Articles ,Cells, Immobilized ,digestive system diseases ,030104 developmental biology ,Cytokines ,Heterografts - Abstract
Human perivascular stem/stromal cells (PSC) are a multipotent mesodermal progenitor cell population defined by their perivascular residence. PSC are most commonly derived from subcutaneous adipose tissue, and recent studies have demonstrated the high potential for clinical translation of this fluorescence-activated cell sorting-derived cell population for bone tissue engineering. Specifically, purified PSC induce greater bone formation than unpurified stroma taken from the same patient sample. In this study, we examined the differences in early innate immune response to human PSC or unpurified stroma (stromal vascular fraction [SVF]) during the in vivo process of bone formation. Briefly, SVF or PSC from the same patient sample were implanted intramuscularly in the hindlimb of severe combined immunodeficient (SCID) mice using an osteoinductive demineralized bone matrix carrier. Histological examination of early inflammatory infiltrates was examined by hematoxylin and eosin and immunohistochemical staining (Ly-6G, F4/80). Results showed significantly greater neutrophilic and macrophage infiltrates within and around SVF in comparison to PSC-laden implants. Differences in early postoperative inflammation among SVF-laden implants were associated with reduced osteogenic differentiation and bone formation. Similar findings were recapitulated with PSC implantation in immunocompetent mice. Exaggerated postoperative inflammation was associated with increased IL-1α, IL-1β, IFN-γ, and TNF-α gene expression among SVF samples, and conversely increased IL-6 and IL-10 expression among PSC samples. These data document a robust immunomodulatory effect of implanted PSC, and an inverse correlation between host inflammatory cell infiltration and stromal progenitor cell-mediated ossification.
- Published
- 2018
- Full Text
- View/download PDF
30. Combining Smoothened Agonist and NEL-Like Protein-1 Enhances Bone Healing
- Author
-
Chenchao Wang, Min Lee, Jia Shen, Swati Shrestha, Chia Soo, Catherine Ding, Soonchul Lee, Aaron W. James, Kang Ting, Carolyn A. Meyers, Eric X. Chen, and Hsin Chuan Pan
- Subjects
Male ,0301 basic medicine ,Agonist ,medicine.medical_specialty ,Bone Regeneration ,medicine.drug_class ,Bone healing ,Temporal bone surgery ,Statistics, Nonparametric ,Fractures, Bone ,Mice ,Random Allocation ,03 medical and health sciences ,Tissue scaffolds ,Osteogenesis ,medicine ,Animals ,Hedgehog Proteins ,Bone regeneration ,Glycoproteins ,Fracture Healing ,Random allocation ,Tissue Scaffolds ,business.industry ,Biopsy, Needle ,Calcium-Binding Proteins ,Temporal Bone ,Combined Modality Therapy ,Immunohistochemistry ,Surgery ,Disease Models, Animal ,030104 developmental biology ,NEL-Like Protein 1 ,Cancer research ,Female ,business ,Smoothened - Abstract
Nonhealing bone defects represent an immense biomedical burden. Despite recent advances in protein-based bone regeneration, safety concerns over bone morphogenetic protein-2 have prompted the search for alternative factors. Previously, the authors examined the additive/synergistic effects of hedgehog and Nel-like protein-1 (NELL-1) on the osteogenic differentiation of mesenchymal stem cells in vitro. In this study, the authors sought to leverage their previous findings by applying the combination of Smoothened agonist (SAG), hedgehog signal activator, and NELL-1 to an in vivo critical-size bone defect model.A 4-mm parietal bone defect was created in mixed-gender CD-1 mice. Treatment groups included control (n = 6), SAG (n = 7), NELL-1 (n = 7), and SAG plus NELL-1 (n = 7). A custom fabricated poly(lactic-co-glycolic acid) disk with hydroxyapatite coating was used as an osteoinductive scaffold.Results at 4 and 8 weeks showed increased bone formation by micro-computed tomographic analyses with either stimulus alone (SAG or NELL-1), but significantly greater bone formation with both components combined (SAG plus NELL-1). This included greater bone healing scores and increased bone volume and bone thickness. Histologic analyses confirmed a significant increase in new bone formation with the combination therapy SAG plus NELL-1, accompanied by increased defect vascularization.In summary, the authors' results suggest that combining the hedgehog signaling agonist SAG and NELL-1 has potential as a novel therapeutic strategy for the healing of critical-size bone defects. Future directions will include optimization of dosage and delivery strategy for an SAG and NELL-1 combination product.
- Published
- 2017
- Full Text
- View/download PDF
31. Vascular patterning in human heterotopic ossification
- Author
-
Benjamin Levi, Aditya Mohan, Aaron W. James, Catherine Ding, Margaret Cocks, Carolyn A. Meyers, and Edward F. McCarthy
- Subjects
Adult ,Male ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Time Factors ,Adolescent ,Angiogenesis ,Biopsy ,Article ,Bone and Bones ,Veins ,Pathology and Forensic Medicine ,Surgical pathology ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Vascularity ,Vasculogenesis ,Osteogenesis ,medicine ,Humans ,Child ,Process (anatomy) ,Aged ,Osteoblasts ,Neovascularization, Pathologic ,business.industry ,Ossification, Heterotopic ,Soft tissue ,Arteries ,Myositis ossificans ,Anatomy ,Middle Aged ,medicine.disease ,Los Angeles ,Capillaries ,030104 developmental biology ,030220 oncology & carcinogenesis ,Baltimore ,Blood Vessels ,Female ,Heterotopic ossification ,medicine.symptom ,business - Abstract
Heterotopic ossification (HO, also termed myositis ossificans) is the formation of extra-skeletal bone in muscle and soft tissues. HO is a tissue repair process gone awry, and is a common complication of surgery and traumatic injury. Medical strategies to prevent and treat HO fall well short of addressing the clinical need. Better characterization of the tissues supporting HO is critical to identifying therapies directed against this common and sometimes devastating condition. The physiologic processes of osteogenesis and angiogenesis are highly coupled and interdependent. However, little efforts have been made to document the vascular patterning within heterotopic ossification. Here, surgical pathology case files of 29 human HO specimens were examined by vascular histomorphometric analysis. Results demonstrate a temporospatial patterning of HO vascularity that depends on the ‘maturity’ of the bony lesion. In sum, human HO demonstrates a time and space dependent pattern of vascularization suggesting a coupled pathophysiologic process involving the coordinate processes of osteogenesis and angiogenesis. Further imaging studies may be used to further characterize vasculogenesis within HO and whether anti-angiogenic therapies are a conceivable future therapy for this common condition.
- Published
- 2017
- Full Text
- View/download PDF
32. Ang-1 and Ang-2 expression in angiomyolipoma and PEComa family tumors
- Author
-
Kang Ting, Michelle A. Scott, Aaron W. James, Sarah M. Dry, Chia Soo, Swati Shrestha, Jia Shen, Paulina Giacomelli, and Carolyn A. Meyers
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Angiomyolipoma ,Lymphangiomyoma ,business.industry ,Cell of origin ,Soft tissue ,medicine.disease ,Angiopoietin ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Antigen ,030220 oncology & carcinogenesis ,cardiovascular system ,medicine ,Immunohistochemistry ,Original Article ,Orthopedics and Sports Medicine ,Pericyte ,business - Abstract
Objective Perivascular epithelioid cell tumors (PEComa) are an uncommon family of soft tissue tumors. Previously, we described that the presence of pericyte antigens among PEComa family tumors differs extensively by histologic appearance. Methods Here, we extend our findings using the pericyte antigens Angiopoietin-1 (Ang-1) and Angiopoietin-2 (Ang-2), using immunohistochemical detection in human tumor samples. Results While Ang-1 showed no expression across any PEComa family tumor, Ang-2 showed expression that like other pericyte markers was largely determined by cytologic appearance. Conclusion/implications Pericytic marker expression in PEComa may represent a true pericytic cell of origin, or alternatively aberrant pericyte marker adoption.
- Published
- 2017
- Full Text
- View/download PDF
33. Human perivascular stem cell-derived extracellular vesicles mediate bone repair
- Author
-
Bruno Péault, Carolyn A. Meyers, Kenneth W. Witwer, Yiyun Wang, Kristen P. Broderick, Ching Yun Hsu, Yongxing Gao, Aaron W. James, Jiajia Xu, Leslie Chang, Catherine Ding, and Leititia Zhang
- Subjects
0301 basic medicine ,Mouse ,QH301-705.5 ,Science ,Exosome ,Osteocytes ,General Biochemistry, Genetics and Molecular Biology ,osteogenesis ,03 medical and health sciences ,Paracrine signalling ,Extracellular Vesicles ,0302 clinical medicine ,pericyte ,medicine ,Humans ,exosome ,Progenitor cell ,Biology (General) ,bone tissue engineering ,Cells, Cultured ,mesenchymal stem cell ,General Immunology and Microbiology ,Chemistry ,General Neuroscience ,Regeneration (biology) ,Stem Cells ,Mesenchymal stem cell ,General Medicine ,Cell Biology ,bone repair ,Stem Cells and Regenerative Medicine ,Coculture Techniques ,Trypsinization ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Blood Vessels ,Medicine ,Pericyte ,Stem cell ,030217 neurology & neurosurgery ,Research Article ,Human - Abstract
The vascular wall is a source of progenitor cells that are able to induce skeletal repair, primarily by paracrine mechanisms. Here, the paracrine role of extracellular vesicles (EVs) in bone healing was investigated. First, purified human perivascular stem cells (PSCs) were observed to induce mitogenic, pro-migratory, and pro-osteogenic effects on osteoprogenitor cells while in non-contact co-culture via elaboration of EVs. PSC-derived EVs shared mitogenic, pro-migratory, and pro-osteogenic properties of their parent cell. PSC-EV effects were dependent on surface-associated tetraspanins, as demonstrated by EV trypsinization, or neutralizing antibodies for CD9 or CD81. Moreover, shRNA knockdown in recipient cells demonstrated requirement for the CD9/CD81 binding partners IGSF8 and PTGFRN for EV bioactivity. Finally, PSC-EVs stimulated bone repair, and did so via stimulation of skeletal cell proliferation, migration, and osteodifferentiation. In sum, PSC-EVs mediate the same tissue repair effects of perivascular stem cells, and represent an ‘off-the-shelf’ alternative for bone tissue regeneration., eLife digest Throughout our lives, our bodies need to heal after injury. Blood vessels are found throughout the body’s tissues and are a source of cells that guide the process of repair. These cells, called perivascular stem cells (PSCs), are a type of stem cell found in the lining of blood vessels. Stem cells are cells that can become one of several different types of mature cells, depending on what the body needs. Extracellular vesicles are bundles of chemical signals that cells send into their external environment. Just like an address or a tag on a parcel, specific molecules mark the exterior surface of these bundles to deliver the message to the right recipient. Stem cells often use extracellular vesicles to communicate with surrounding cells. One role of PSCs is repairing damage to bones. Unusually, they do not turn into new bone cells and so do not directly contribute to the re-growing tissue. Instead, PSCs act indirectly, by stimulating the cells around them. How PSCs send these ‘repair instructions’ has, however, remained unclear. Xu et al. wanted to determine if PSCs used extracellular vesicles to direct bone repair, and if so, what ‘tags’ needed to be on the vesicles and on the receiving cells for this to happen. Experiments using PSCs and immature bone cells grown in the laboratory allowed the PSCs’ effect on bone cells to be simulated in a Petri dish. The two types of cells were grown on either side of a barrier, which separated them physically but allowed chemical signals through. In response to the PSCs, the immature bone cells multiplied, started to move (which is something they need to do to heal damaged tissue), and began to resemble mature bone cells. Analysis of the signals released by the PSCs revealed that these were indeed extracellular vesicles, and that they were tagged by specific proteins called tetraspanins. Genetic manipulation of the immature bone cells later showed that these cells needed specific ‘receiver’ molecules to respond to the PSCs. Adding only extracellular vesicles to the bone cells, without any PSCs, confirmed that it was indeed the vesicles that triggered the healing response. Finally, giving the vesicles to mice with bone damage helped them to heal faster than untreated animals. These results have uncovered a key mechanism by which stem cells control the repair of bone tissue. This could one day lead to better treatments for patients recovering from fractures or needing bone surgery.
- Published
- 2019
34. Author response: Human perivascular stem cell-derived extracellular vesicles mediate bone repair
- Author
-
Leslie Chang, Yiyun Wang, Carolyn A. Meyers, Kenneth W. Witwer, Aaron W. James, Jiajia Xu, Yongxing Gao, Kristen P. Broderick, Leititia Zhang, Bruno Péault, Ching Yun Hsu, and Catherine Ding
- Subjects
Chemistry ,Bone healing ,Stem cell ,Extracellular vesicles ,Cell biology - Published
- 2019
- Full Text
- View/download PDF
35. Differential Vascularity in Genetic and Nonhereditary Heterotopic Ossification
- Author
-
Alisha D. Ware, Eileen M. Shore, Edward F. McCarthy, Aaron W. James, Carol D. Morris, Carolyn A. Meyers, and Niambi Brewer
- Subjects
0301 basic medicine ,Adult ,Male ,Pathology ,medicine.medical_specialty ,Angiogenesis ,Biopsy ,Progressive osseous heteroplasia ,Bone and Bones ,Pathology and Forensic Medicine ,Diagnosis, Differential ,03 medical and health sciences ,0302 clinical medicine ,Vascularity ,Spatio-Temporal Analysis ,medicine ,Humans ,Child ,business.industry ,Ossification, Heterotopic ,Skin Diseases, Genetic ,Heterotopic bone ,medicine.disease ,Bone Diseases, Metabolic ,030104 developmental biology ,medicine.anatomical_structure ,Myositis Ossificans ,030220 oncology & carcinogenesis ,Fibrodysplasia ossificans progressiva ,Child, Preschool ,Mutation ,Wounds and Injuries ,Surgery ,Heterotopic ossification ,Anatomy ,medicine.symptom ,business ,Complication ,Blood vessel - Abstract
Introduction. Nonhereditary heterotopic ossification (NHO) is a common complication of trauma. Progressive osseous heteroplasia (POH) and fibrodysplasia ossificans progressiva (FOP) are rare genetic causes of heterotopic bone. In this article, we detail the vascular patterning associated with genetic versus NHO. Methods. Vascular histomorphometric analysis was performed on patient samples from POH, FOP, and NHO. Endpoints for analysis included blood vessel (BV) number, area, density, size, and wall thickness. Results. Results demonstrated conserved temporal dynamic changes in vascularity across all heterotopic ossification lesions. Immature areas had the highest BV number, while the more mature foci had the highest BV area. Most vascular parameters were significantly increased in genetic as compared with NHO. Discussion. In sum, both genetic and NHO show temporospatial variation in vascularity. These findings suggest that angiogenic pathways are potential therapeutic targets in both genetic and nonhereditary forms of heterotopic ossification.
- Published
- 2019
36. Abstract 104: Identification of the Role of Sensory Nerve TrkA Signaling on Progenitor Cell Fate after Extremity Trauma
- Author
-
Paul S. Cederna, Seungyong Lee, Stephen W.P. Kemp, Aaron W. James, Thomas L. Clemens, Zaid N. Khatib, Talis Rehse, Chase A. Pagani, Jeffrey Lisiecki, Carrie A. Kubiak, Carolyn A. Meyers, Michael Sorkin, Mohamed A. Garada, Noelle D. Visser, Benjamin Levi, Prasanth Kotha, Charles Hwang, Kaetlin Vasquez, and David M. Stepien
- Subjects
medicine.anatomical_structure ,PSRC 2019 Abstract Supplement ,business.industry ,lcsh:Surgery ,Medicine ,Surgery ,Identification (biology) ,lcsh:RD1-811 ,Tropomyosin receptor kinase A ,Progenitor cell ,business ,Neuroscience ,Sensory nerve - Published
- 2019
37. Relative contributions of adipose-resident CD146 pericytes and CD34 adventitial progenitor cells in bone tissue engineering
- Author
-
Carolyn A. Meyers, Yiyun Wang, Aaron W. James, Lei Zhang, Jiajia Xu, Kristen P. Broderick, Leslie Chang, Min Lee, and Bruno Péault
- Subjects
0301 basic medicine ,Cell type ,Stromal cell ,Angiogenesis ,Chemistry ,Mesenchymal stem cell ,lcsh:R ,Biomedical Engineering ,CD34 ,Medicine (miscellaneous) ,Connective tissue ,lcsh:Medicine ,Cell Biology ,Article ,3. Good health ,Cell biology ,Cell therapy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,medicine ,Progenitor cell ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Pericytes and other perivascular stem/stromal cells are of growing interest in the field of tissue engineering. A portion of perivascular cells are well recognized to have MSC (mesenchymal stem cell) characteristics, including multipotentiality, self-renewal, immunoregulatory functions, and diverse roles in tissue repair. Here, we investigate the differential but overlapping roles of two perivascular cell subsets in paracrine induction of bone repair. CD146+CD34−CD31−CD45−pericytes and CD34+CD146−CD31−CD45−adventitial cells were derived from human adipose tissue and applied alone or in combination to calvarial bone defects in mice. In vitro, osteogenic differentiation and tubulogenesis assays were performed using either fluorescence activated cell sorting-derived CD146+ pericytes or CD34+ adventitial cells. Results showed that CD146+ pericytes induced increased cord formation in vitro and angiogenesis in vivo in comparison with patient-matched CD34+ adventitial cells. In contrast, CD34+ adventitial cells demonstrated heightened paracrine-induced osteogenesis in vitro. When applied in a critical-size calvarial defect model in NOD/SCID mice, the combination treatment of CD146+ pericytes with CD34+ adventitial cells led to greater re-ossification than either cell type alone. In summary, adipose-derived CD146+ pericytes and CD34+ adventitial cells display functionally distinct yet overlapping and complementary roles in bone defect repair. Consequently, CD146+ pericytes and CD34+ adventitial cells may demonstrate synergistic bone healing when applied as a combination cellular therapy., Bone repair: synergistic healing from two types of fat cells Different kinds of cells found surrounding blood vessels in fat play a complementary and synergistic role in bone healing. Aaron James from Johns Hopkins University in Baltimore, MD, USA, and colleagues derived two subsets of cells from human fat tissue: contractile cells known as pericytes that wrap around cellular lining of capillaries and tiny veins; and connective tissue cells known as adventitial cells that surrounds larger vessels. Under isolated culture conditions, pericytes stimulated the development of primitive blood vessels, whereas adventitial cells promoted early bone formation. The researchers applied the cells to the sites of bone defects in mice and saw that combined treatment with both pericytes and adventitial cells led to greater bone repair than treatment with either cell type alone.
- Published
- 2019
- Full Text
- View/download PDF
38. Perivascular Progenitor Cells for Bone Regeneration
- Author
-
Winters R. Hardy, Noah Yan, Kang Ting, Aaron W. James, Hindle P, Bruno Péault, Chia Soo, Jia Jia Xu, Carolyn A. Meyers, Greg Asatrian, and Kristen P. Broderick
- Subjects
education.field_of_study ,Pathology ,medicine.medical_specialty ,Stromal cell ,Regeneration (biology) ,Mesenchymal stem cell ,Population ,Biology ,medicine.anatomical_structure ,medicine ,Pericyte ,Progenitor cell ,Bone regeneration ,education ,Multipotentiality - Abstract
Perivascular progenitor cells are of growing interest in the field of bone tissue engineering. Perivascular progenitor cells have mesenchymal stem/stromal cell (MSC) characteristics, including multipotentiality, self-renewal, immunomodulatory functions, and diverse roles in tissue repair. From human tissue, the purification of perivascular progenitor cells is most common from subcutaneous white adipose tissue, although all vascularized organs studied to date have a perivascular progenitor cell population. Microvascular pericytes are commonly isolated as a CD146+CD34−CD31−CD45− cell population, while adventitial progenitor cells are more commonly identified as a CD146−CD34+CD31−CD45− population. Perivascular progenitor cells have been applied in diverse orthopedic conditions, including both ectopic and orthotopic models of bone formation/regeneration. This review covers studies to date in bone tissue engineering as well as several emerging areas of study, including the concept of regional specification within the perivascular niche.
- Published
- 2019
- Full Text
- View/download PDF
39. Pericytes in Sarcomas and Other Mesenchymal Tumors
- Author
-
Leslie Chang, Michelle A. Scott, Aaron W. James, and Carolyn A. Meyers
- Subjects
Pathology ,medicine.medical_specialty ,Myofibroma ,Myopericytoma ,Liposarcoma ,Biology ,medicine.disease ,Mural cell ,Glomus tumor ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,medicine ,CD146 ,030212 general & internal medicine ,Pericyte ,Sarcoma - Abstract
Tumors of mesenchymal origin are a diverse group, with >130 distinct entities currently recognized by the World Health Organization. A subset of mesenchymal tumors grow or invade in a perivascular fashion, and their potential relationship to pericytes is a matter of ongoing interest. In fact, multiple intersections exist between pericytes and tumors of mesenchymal origin. First, pericytes are the likely cell of origin for a group of mesenchymal tumors with a common perivascular growth pattern. These primarily benign tumors grow in a perivascular fashion and diffusely express canonical pericyte markers such as CD146, smooth muscle actin (SMA), platelet-derived growth factor receptor beta (PDGFR-β), and RGS5. These benign tumors include glomus tumor, myopericytoma, angioleiomyoma, and myofibroma. Second and as suggested by animal models, pericytes may give rise to malignant sarcomas. This is not a suggestion that all sarcomas within a certain subtype arise from pericytes, but that genetic modifications within a pericyte cell type may give rise to sarcomas. Third, mesenchymal tumors that are likely not a pericyte derivative co-opt pericyte markers in certain contexts. These include the PEComa family of tumors and liposarcoma. Fourth and finally, as “guardians” that enwrap the microvasculature, nonneoplastic pericytes may be important in sarcoma disease progression.
- Published
- 2019
- Full Text
- View/download PDF
40. Sclerostin expression in skeletal sarcomas
- Author
-
Jia Shen, Fritz C. Eilber, Swati Shrestha, Chia Soo, Noah Federman, Vi Nguyen, Carolyn A. Meyers, Aaron W. James, Arun S. Singh, Greg Asatrian, Kang Ting, Sarah M. Dry, Greg LaChaud, and Nicholas M. Bernthal
- Subjects
0301 basic medicine ,Aging ,Pathology ,Biopsy ,Enchondroma ,Osteoporosis ,chemistry.chemical_compound ,0302 clinical medicine ,Osteogenesis ,Neoplasms ,Wnt Signaling Pathway ,Cancer ,Osteosarcoma ,Tumor ,Wnt signaling pathway ,Adaptor Proteins ,Cell Differentiation ,Sarcoma ,Immunohistochemistry ,Phenotype ,030220 oncology & carcinogenesis ,Bone Morphogenetic Proteins ,SOST ,Genetic Markers ,medicine.medical_specialty ,Osteoma, Osteoid ,Bone Tissue ,Clinical Sciences ,Chondrosarcoma ,Neoplasms, Bone Tissue ,Bone Neoplasms ,Biology ,Article ,Cell Line ,Pathology and Forensic Medicine ,03 medical and health sciences ,Cell Line, Tumor ,Biomarkers, Tumor ,Genetics ,medicine ,Humans ,Osteoblastoma ,Adaptor Proteins, Signal Transducing ,Retrospective Studies ,Osteoid ,Signal Transducing ,Osteoma ,Stem Cell Research ,Alkaline Phosphatase ,medicine.disease ,Wnt signaling ,030104 developmental biology ,chemistry ,Musculoskeletal ,Sclerostin ,Chondroma ,Biomarkers - Abstract
Sclerostin (SOST) is an extracellular Wnt signaling antagonist which negatively regulates bone mass. Despite this, the expression and function of SOST in skeletal tumors remain poorly described. Here, we first describe the immunohistochemical staining pattern of SOST across benign and malignant skeletal tumors with bone or cartilage matrix (n = 68 primary tumors). Next, relative SOST expression was compared to markers of Wnt signaling activity and osteogenic differentiation across human osteosarcoma (OS) cell lines (n = 7 cell lines examined). Results showed immunohistochemical detection of SOST in most bone-forming tumors (90.2%; 46/51) and all cartilage-forming tumors (100%; 17/17). Among OSs, variable intensity and distribution of SOST expression were observed, which highly correlated with the presence and degree of neoplastic bone. Patchy SOST expression was observed in cartilage-forming tumors, which did not distinguish between benign and malignant tumors or correlate with regional morphologic characteristics. Finally, SOST expression varied widely between OS cell lines, with more than 97-fold variation. Among OS cell lines, SOST expression positively correlated with the marker of osteogenic differentiation alkaline phosphatase and did not correlate well with markers of Wnt/β-catenin signaling activity. In summary, SOST is frequently expressed in skeletal bone- and cartilage-forming tumors. The strong spatial correlation with bone formation and the in vitro expression patterns are in line with the known functions of SOST in nonneoplastic bone, as a feedback inhibitor on osteogenic differentiation. With anti-SOST as a potential therapy for osteoporosis in the near future, its basic biologic and phenotypic consequences in skeletal tumors should not be overlooked.
- Published
- 2016
- Full Text
- View/download PDF
41. Basic and Translational Research in Osteosarcoma: A Year in Review
- Author
-
Le Chang, Michelle A. Scott, Aaron W. James, Carolyn A. Meyers, Jia Shen, and Greg Asatrian
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Internal medicine ,Year in review ,medicine ,Osteosarcoma ,Translational research ,business ,medicine.disease - Published
- 2016
- Full Text
- View/download PDF
42. The Role of Integrins in Osteosarcoma
- Author
-
Michelle A. Scott, Vi Nguyen, Aaron W. James, Alan Nguyen, Jia Shen, and Carolyn A. Meyers
- Subjects
0301 basic medicine ,biology ,business.industry ,Integrin ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Medicine ,Osteosarcoma ,business - Published
- 2016
- Full Text
- View/download PDF
43. Pericytes for Therapeutic Bone Repair
- Author
-
Lei Zhang, Leslie Chang, Joan Casamitjana, Bruno Péault, Aaron W. James, and Carolyn A. Meyers
- Subjects
0301 basic medicine ,Bone Regeneration ,Angiogenesis ,Adipose tissue ,Calvaria ,Bone healing ,Article ,03 medical and health sciences ,Mice ,Dogs ,Osteogenesis ,medicine ,Animals ,Bone regeneration ,Tissue Engineering ,Chemistry ,Mesenchymal stem cell ,Cell Differentiation ,Mesenchymal Stem Cells ,Cell biology ,Rats ,030104 developmental biology ,medicine.anatomical_structure ,Pericyte ,Stem cell ,Pericytes - Abstract
Besides seminal functions in angiogenesis and blood pressure regulation, microvascular pericytes possess a latent tissue regenerative potential that can be revealed in culture following transition into mesenchymal stem cells. Endowed with robust osteogenic potential, pericytes and other related perivascular cells extracted from adipose tissue represent a potent and abundant cell source for refined bone tissue engineering and improved cell therapies of fractures and other bone defects. The use of diverse bone formation assays in vivo, which include mouse muscle pocket osteogenesis and calvaria replenishment, rat and dog spine fusion, and rat non-union fracture healing, has confirmed the superiority of purified perivascular cells for skeletal (re)generation. As a surprising observation though, despite strong endogenous bone-forming potential, perivascular cells drive bone regeneration essentially indirectly, via recruitment by secreted factors of local osteo-progenitors.
- Published
- 2018
44. Overlapping features of rapidly progressive osteoarthrosis and Charcot arthropathy
- Author
-
Daniel Miller, Catherine Ding, Takashi Sono, Carolyn A. Meyers, Aaron W. James, and Edward F. McCarthy
- Subjects
musculoskeletal diseases ,030222 orthopedics ,Pathology ,medicine.medical_specialty ,Joint destruction ,biology ,business.industry ,030229 sport sciences ,medicine.disease ,Skeleton (computer programming) ,Article ,03 medical and health sciences ,0302 clinical medicine ,Nerve growth factor ,nervous system ,Diabetes mellitus ,Arthropathy ,Neuropathic arthropathy ,biology.protein ,Medicine ,Orthopedics and Sports Medicine ,Syphilis ,business ,Neurotrophin - Abstract
Nerve growth factor (NGF) is the primary neurotrophin in the skeleton and a central mediator of skeletal pain. Recent trials of anti-NGF neutralizing antibodies have resulted in infrequent but well-described incidence of rapidly progressive osteoarthrosis (RPOA). Neuropathy, whether from syphilis or diabetes, is also associated with severe joint destruction, known as neuroarthropathy or Charcot joint. These commonalities of severe joint destruction with either loss of a neurotrophin (anti-NGF) or a deficit of functional skeletal innervation led us to examine our institutional case files for potential radio-pathologic overlap between RPOA and Charcot joint.
- Published
- 2018
45. Age dependent effects of NELL-1 isoforms on bone marrow stromal cells
- Author
-
Amy Lu, Catherine Ding, Aaron W. James, Carolyn A. Meyers, Leslie Chang, Shen Pang, Zhibo Sun, and Kang Ting
- Subjects
chemistry.chemical_classification ,Gene isoform ,030222 orthopedics ,Stromal cell ,Ossification ,business.industry ,Mesenchymal stem cell ,030229 sport sciences ,Cell morphology ,Molecular biology ,Article ,Amino acid ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,chemistry ,Gene expression ,medicine ,Orthopedics and Sports Medicine ,Bone marrow ,medicine.symptom ,business - Abstract
Objective NELL-1 is an osteogenic protein first discovered to control ossification of the cranium. NELL-1 exists in at least two isoforms. The full-length NELL-1 contains 810 amino acid (aa) (NELL-1810), the N-terminal-truncated NELL-1 isoform contains 570 aa (NELL-1570). The differences in cellular effects between NELL-1 isoforms are not well understood. Methods: Here, BMSC were derived from adult or aged mice, followed by overexpression of NELL-1810 or NELL-1570. Cell morphology, proliferation, and gene expression were examined. Results/Conclusions: Overall, the proliferative effect of NELL-1570 was age dependent, showing prominent induction in adult but not aged mice.
- Published
- 2018
46. Heterotopic Ossification: A Comprehensive Review
- Author
-
Jeffrey Lisiecki, Aaron W. James, Benjamin Levi, Laura M. Fayad, Catherine Ding, Edward F. McCarthy, Sarah Miller, Carolyn A. Meyers, Takashi Sono, and Adam S. Levin
- Subjects
030222 orthopedics ,0303 health sciences ,business.industry ,Endocrinology, Diabetes and Metabolism ,Soft tissue ,Heterotopic bone ,Review ,Myositis ossificans ,Tissue repair ,medicine.disease ,Bioinformatics ,HETEROTOPIC BONE ,Ectopic bone formation ,FIBRODYSPLASIA OSSIFICANS PROGRESSIVA ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,Fibrodysplasia ossificans progressiva ,medicine ,MYOSITIS OSSIFICANS ,Orthopedics and Sports Medicine ,Heterotopic ossification ,business ,ECTOPIC BONE ,030304 developmental biology - Abstract
Heterotopic ossification (HO) is a diverse pathologic process, defined as the formation of extraskeletal bone in muscle and soft tissues. HO can be conceptualized as a tissue repair process gone awry and is a common complication of trauma and surgery. This comprehensive review seeks to synthesize the clinical, pathoetiologic, and basic biologic features of HO, including nongenetic and genetic forms. First, the clinical features, radiographic appearance, histopathologic diagnosis, and current methods of treatment are discussed. Next, current concepts regarding the mechanistic bases for HO are discussed, including the putative cell types responsible for HO formation, the inflammatory milieu and other prerequisite “niche” factors for HO initiation and propagation, and currently available animal models for the study of HO of this common and potentially devastating condition. © 2019 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
- Published
- 2018
47. Effects of WNT3A and WNT16 on the Osteogenic and Adipogenic Differentiation of Perivascular Stem/Stromal Cells
- Author
-
Chia Soo, Swati Shrestha, Greg Asatrian, Xuepeng Chen, Haichao Jia, Aaron W. James, Rebecca Tsuei, Catherine Ding, Xinli Zhang, Jia Shen, Bruno Péault, Kang Ting, and Carolyn A. Meyers
- Subjects
0301 basic medicine ,Stromal cell ,animal structures ,endocrine system diseases ,Population ,Cell ,Biomedical Engineering ,Bioengineering ,Biology ,Biochemistry ,digestive system ,Biomaterials ,03 medical and health sciences ,Osteogenesis ,Wnt3A Protein ,medicine ,Humans ,education ,Wnt Signaling Pathway ,Cells, Cultured ,education.field_of_study ,Adipogenesis ,Tissue Engineering ,digestive, oral, and skin physiology ,Wnt signaling pathway ,Cell Differentiation ,Mesenchymal Stem Cells ,Original Articles ,Cell sorting ,digestive system diseases ,Wnt Proteins ,030104 developmental biology ,medicine.anatomical_structure ,DKK1 ,Cancer research ,Stromal Cells ,WNT3A - Abstract
Human perivascular stem/stromal cells (hPSC) are a multipotent mesenchymogenic stromal cell population defined by their perivascular locale. Recent studies have demonstrated the high potential for clinical translation of this fluorescence-activated cell sorting (FACS)-derived cell population for autologous bone tissue engineering. However, the mechanisms underlying the osteogenic differentiation of PSC are incompletely understood. The current study investigates the roles of canonical and noncanonical Wnt signaling in the osteogenic and adipogenic differentiation of PSC. Results showed that both canonical and noncanonical Wnt signaling activity transiently increased during PSC osteogenic differentiation in vitro. Sustained WNT3A treatment significantly decreased PSC osteogenic differentiation. Conversely, sustained treatment with Wnt family member 16 (WNT16), a mixed canonical and noncanonical ligand, increased osteogenic differentiation in a c-Jun N-terminal kinase (JNK) pathway-dependent manner. Conversely, WNT16 knockdown significantly diminished PSC osteogenic differentiation. Finally, WNT16 but not WNT3A increased the adipogenic differentiation of PSC. These results indicate the importance of regulation of canonical and noncanonical Wnt signaling for PSC fate and differentiation. Moreover, these data suggest that WNT16 plays a functional and necessary role in PSC osteogenesis.
- Published
- 2018
48. NELL-1 induces Sca-1+ mesenchymal progenitor cell expansion in models of bone maintenance and repair
- Author
-
Chia Soo, Carolyn A. Meyers, Jin H. Kwak, Aaron W. James, Alan Nguyen, Greg Asatrian, Jia Shen, Kevork Khadarian, Xinli Zhang, Rebecca Tsuei, Cymbeline T. Culiat, Min Lee, Weiming Li, Hsin Chuan Pan, and Kang Ting
- Subjects
0301 basic medicine ,Ossification ,Mesenchymal stem cell ,Wnt signaling pathway ,General Medicine ,Biology ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,medicine ,Cancer research ,Systemic administration ,Bone marrow ,medicine.symptom ,Progenitor cell ,Stem cell ,Haploinsufficiency ,Research Article - Abstract
NELL-1 is a secreted, osteogenic protein first discovered to control ossification of the cranial skeleton. Recently, NELL-1 has been implicated in bone maintenance. However, the cellular determinants of NELL-1's bone-forming effects are still unknown. Here, recombinant human NELL-1 (rhNELL-1) implantation was examined in a clinically relevant nonhuman primate lumbar spinal fusion model. Prolonged rhNELL-1 protein release was achieved using an apatite-coated β-tricalcium phosphate carrier, resulting in a local influx of stem cell antigen-1-positive (Sca-1+) mesenchymal progenitor cells (MPCs), and complete osseous fusion across all samples (100% spinal fusion rate). Murine studies revealed that Nell-1 haploinsufficiency results in marked reductions in the numbers of Sca-1+CD45-CD31- bone marrow MPCs associated with low bone mass. Conversely, rhNELL-1 systemic administration in mice showed a marked anabolic effect accompanied by increased numbers of Sca-1+CD45-CD31- bone marrow MPCs. Mechanistically, rhNELL-1 induces Sca-1 transcription among MPCs, in a process requiring intact Wnt/β-catenin signaling. In summary, NELL-1 effectively induces bone formation across small and large animal models either via local implantation or intravenous delivery. NELL-1 induces an expansion of a bone marrow subset of MPCs with Sca-1 expression. These findings provide compelling justification for the clinical translation of a NELL-1-based therapy for local or systemic bone formation.
- Published
- 2017
- Full Text
- View/download PDF
49. Isolation and characterization of canine perivascular stem/stromal cells for bone tissue engineering
- Author
-
Mihaela Crisan, Greg Asatrian, Erin Zou, Venu Lagishetty, Winters R. Hardy, Kang Ting, Xinli Zhang, Pei Liang, Sonja Lobo, Catherine Ding, Martin K. Childers, Carolyn A. Meyers, Bruno Péault, Yu Hsin Yen, Aaron W. James, and Chia Soo
- Subjects
0301 basic medicine ,Vascular Endothelial Growth Factor A ,Pathology ,Physiology ,Cellular differentiation ,CD34 ,Adipose tissue ,lcsh:Medicine ,Alizarin Staining ,Cell Separation ,Spectrum Analysis Techniques ,Endocrinology ,Tissue engineering ,Osteogenesis ,Animal Cells ,Medicine and Health Sciences ,Group-Specific Staining ,Enzyme-Linked Immunoassays ,lcsh:Science ,Cells, Cultured ,Platelet-Derived Growth Factor ,Staining ,Fluorescence-Activated Cell Sorting ,Multidisciplinary ,Stem Cells ,digestive, oral, and skin physiology ,Cell Differentiation ,Recombinant Proteins ,Adipose Tissue ,Spectrophotometry ,CD146 ,Engineering and Technology ,Fibroblast Growth Factor 2 ,Cytophotometry ,Anatomy ,Cellular Types ,Research Article ,Biotechnology ,medicine.medical_specialty ,Stromal cell ,Nerve Tissue Proteins ,Bioengineering ,Biology ,Research and Analysis Methods ,Bone and Bones ,03 medical and health sciences ,Dogs ,Growth Factors ,medicine ,Journal Article ,Animals ,Humans ,Progenitor cell ,Immunoassays ,Tissue Engineering ,Endocrine Physiology ,Mesenchymal stem cell ,lcsh:R ,Calcium-Binding Proteins ,Biology and Life Sciences ,Mesenchymal Stem Cells ,Cell Biology ,digestive system diseases ,030104 developmental biology ,Biological Tissue ,Specimen Preparation and Treatment ,Immunologic Techniques ,lcsh:Q ,Stromal Cells ,Pericytes - Abstract
For over 15 years, human subcutaneous adipose tissue has been recognized as a rich source of tissue resident mesenchymal stem/stromal cells (MSC). The isolation of perivascular progenitor cells from human adipose tissue by a cell sorting strategy was first published in 2008. Since this time, the interest in using pericytes and related perivascular stem/stromal cell (PSC) populations for tissue engineering has significantly increased. Here, we describe a set of experiments identifying, isolating and characterizing PSC from canine tissue (N = 12 canine adipose tissue samples). Results showed that the same antibodies used for human PSC identification and isolation are cross-reactive with canine tissue (CD45, CD146, CD34). Like their human correlate, canine PSC demonstrate characteristics of MSC including cell surface marker expression, colony forming unit-fibroblast (CFU-F) inclusion, and osteogenic differentiation potential. As well, canine PSC respond to osteoinductive signals in a similar fashion as do human PSC, such as the secreted differentiation factor NEL-Like Molecule-1 (NELL-1). Nevertheless, important differences exist between human and canine PSC, including differences in baseline osteogenic potential. In summary, canine PSC represent a multipotent mesenchymogenic cell source for future translational efforts in tissue engineering.
- Published
- 2017
- Full Text
- View/download PDF
50. BMP-2-induced bone formation and neural inflammation
- Author
-
Carolyn A. Meyers, Vi Nguyen, Shailesh Agarwal, Aaron W. James, Benjamin Levi, and Noah Yan
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,biology ,business.industry ,medicine.medical_treatment ,Neuritis ,Inflammation ,Context (language use) ,Transforming growth factor beta ,Bone morphogenetic protein ,Bone morphogenetic protein 2 ,Article ,Bone morphogenetic protein 7 ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Cytokine ,medicine ,biology.protein ,Cancer research ,Orthopedics and Sports Medicine ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Bone morphogenetic protein-2 (BMP-2), a potent osteoinductive cytokine from the transforming growth factor beta (TGF-β) family, is currently the most commonly used protein-based bone graft substitute. Although clinical use of BMP-2 has significantly increased in recent years, its prominence has also highlighted various adverse events, including induction of inflammation. This review will elucidate the relationship between BMP-2 and inflammation, with an emphasis on peripheral nerve inflammation and its sequelae. As well, we review the potential additive roles of nerve released factors with BMP2 in the context of bone formation.
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.